Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR INHIBITING AND TREATING NEUROLOGICAL CONDITIONS
Document Type and Number:
WIPO Patent Application WO/2016/036403
Kind Code:
A1
Abstract:
This document provides methods and materials related to treating subjects having specific genetic variations associated with neurological disorders such as Parkinson's disease.

Inventors:
HATCHWELL ELI (GB)
EIS PEGGY (US)
Application Number:
PCT/US2015/000093
Publication Date:
March 10, 2016
Filing Date:
September 04, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
POPULATION DIAGNOSTICS INC (US)
International Classes:
A61K31/015; C12Q1/68; A61K31/122; A61K31/355; A61K31/375; A61K31/385; A61K31/51; A61K31/7105; A61K33/00; A61P25/28
Foreign References:
US20140162933A12014-06-12
US20120059594A12012-03-08
Other References:
EIS ET AL.: "Genetic variation in mitochondrial complex I genes are associated with Parkinson' s disease", KEYSTONE SYMPOSIUM POSTER PRESENTATION: PARKINSON'S DISEASE: GENETICS, MECHANISMS AND THERAPEUTICS, 3 March 2014 (2014-03-03)
NISHIOKA ET AL.: "Genetic variation of the mitochondrial complex I subunit NDUFV2 and Parkinson's disease", PARKINSONISM AND RELATED DISORDERS, vol. 16, no. 10, 2010, pages 686 - 687, XP027539381
CALVO ET AL.: "High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency", NATURE GENETICS, vol. 42, no. 10, 2010, pages 851 - 858, XP055244080, DOI: doi:10.1038/ng.659
Attorney, Agent or Firm:
PERDOK, Monique, M. et al. (Minneapolis, MN, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method of diagnosing a susceptibility to a movement disorder such as Parkinson's Disease in a subject, comprising: providing nucleic acid sequence information from the subject on the presence or absence of at least genetic variation in one or more genes or regions in Figure 8A or Figure 8D; and diagnosing a susceptibility to the movement disoder in the subject if the subject has at least one genetic variation in the one or more genes or regions in Figure 8A or Figure 8D, wherein the at least genetic variation occurs in the gene or region more frequently in a population of subjects that have movement disorder than in a population of subjects that does not have movement disorder.

2. The method of claim 1 further comprising assaying a nucleic acid sample of the subject thereby providing the nucleic acid sequence information.

3. The method of claim 1 or 2 wherein the at least one genetic variation is a copy number variation in a subregion in Figure 9A or Figure 9D.

4. The method of any one of claims 1 to 3 wherein the sample is from blood, saliva, urine, serum, tears, skin, tissue, or hair.

5. The method of any one of claims 1 to 4 wherein the sample comprises purified nucleic acid. 6. The method of any one of claims 1 to 5 wherein the sample comprises amplified nucleic acid.

7. The method of any one of claims 1 to 6 wherein a microarray analysis is employed to provide the nucleic acid sequence information.

8. The method of claim 7 wherein the microarray analysis comprises a CGH array analysis or a SNP microarray analysis.

9. The method of claim 1 or 2 wherein the nucleic acid information is obtained by an amplification reaction, sequencing, Northern blots, or any combination thereof.

10. The method of any one of claims 1 to 9 wherein the diagnosis includes a neurological examination and/or medical history analysis of the subject.

11. The method of any one of claims 1 to 10 wherein the nucleic acid sequence information or the at least one genetic variation is compared to those of subjects that do not have the movement disorder or are not suspected of having the movement disorder. 12. The method of any one of claims 1 to 11 wherein the at least one genetic variation comprises one or more point mutations, SNPs, SNVs, polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof.

13. The method of any one of claims 1 to 12 wherein the genetic variation comprises one or more CNVs that disrupt or modulate one or more genes listed in Figure 10A, Figure 10D, Figure 11 A or Figure 11 D. 14. The method of any one of claims 1 to 13 wherein the at least one genetic variation comprises one or more CNVs that disrupt or modulate the expression or function of one or more RNA transcripts in Figure 11 A or Figure 11 D.

15. The method of any one of claims 1 to 14 wherein the nucleic acid sequencing information is obtained for the whole genome or whole exome from the one or more subjects.

16. The method of any one of claims 1 to 15 wherein the nucleic acid sequencing information is obtained for the whole genome or whole exome from the one or more subjects and the nucleic acid information is obtained from in silico analysis.

17. The method of any one of claims 1 to 16 wherein the subject is asymptomatic.

18. The method of any one of claims 1 to 17 wherein the genetic variation is in LYG1, L YG2, SUMF1, GNS/RASSF3, ARSB, GALNS, or PSAP.

19. A method of screening a subject for a genetic variation that disrupts or modulates one or more genes in Figure 10A or Figure 10D, comprising: assaying a nucleic acid sample obtained from the subject for a genetic variation in one or more genes in Figure 10A or Figure 10D, wherein the genetic variation is associated with a neurological or movement disorder or an altered susceptibility to a neurological or movement disorder, and detecting genetic variations that disrupt or modulate the one or more genes. 20. The method of claim 19 wherein the genetic variation is associated with Parkinson's Disease or an altered susceptibility to Parkinson's Disease (PD).

21. The method of claim 19 or 20 wherein the genetic variation is in one of SEQ ID Nos:1 to 197 or 806 to 916 or sub-regions in Figure 9A or Figure 9D.

22. The method of any one of claims 19 to 21 wherein the genetic variation comprises one or more point mutations, polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), single nucleotide polymorphisms (SNPs), single nucleotide variations (SNVs), or any combination thereof, wherein the genetic variation results in a loss of gene function or a gain of gene function of the one or more genes in Figure 11 A or Figure 11 D.

23. The method of claim 19 wherein the genetic variation disrupts or modulates a genomic sequence encoded by any one of SEQ ID Nos 198-805, or an RNA transcript or polypeptide expressed therefrom.

24. The method of any one of claims 19 to 23 wherein the assaying comprises detecting nucleic acid information from the nucleic acid sample by one or more methods selected from the group consisting of PCR, sequencing, a Northern blot, amplification, hybridization, microarray analysis, or any combination thereof.

25. The method of claim 24 wherein the assaying is sequencing, and the sequencing comprises a sequencing method selected from the group consisting of Massively Parallel Signature Sequencing (MPSS), polony sequencing, 454 pyrosequencing, lllumina sequencing, SOLiD sequencing, ion semiconductor sequencing, DNA nanoball sequencing, heliscope single molecule sequencing, single molecule real time (SMRT) sequencing, RNAP sequencing, Nanopore DNA sequencing, sequencing by hybridization, or microfluidic Sanger sequencing.

26. The method of any one of claims 19 to 25 wherein the nucleic acid sample is collected from blood, saliva, urine, serum, tears, skin, tissue, or hair from the subject.

27. The method of any one of claims 19 to 26 wherein the assaying the nucleic acid sample from the subject comprises purifying polynucleotides from the nucleic acid sample; and directly assaying one or more of the purified polynucleotides, amplifying a nucleotide sequence of the purified polynucleotides, or performing a microarray analysis of the purified polynucleotides.

28. The method of claim 24 wherein the assaying is microarray analysis, and wherein the microarray analysis comprises a Comparative Genomic Hybridization (CGH) array analysis or an SNP array analysis.

29. The method of any one of claims 1 to 18 further comprising providing an evaluation of the subject's motor skills, autonomic function, neuropsychiatry, mood, cognition, behavior, thoughts, ability to sense, family history of early-onset neurological disorders or late-onset neurological disorders, or a combination thereof.

30. The method of claim 20 further comprising selecting one or more therapies based on the presence of the genetic variation and administering the one or more therapies.

31. The method of any one of claims 19 to 28 or 30 wherein the assaying the nucleic acid sample obtained from the subject comprises analyzing the whole genome or whole exome of the subject. 32. The method of any one of claims 19 to 23 or 30 to 31 wherein nucleic acid information has already been obtained for the whole genome or whole exome of the subject and the nucleic acid information is obtained from in silico analysis.

33. The method of claim 19 wherein the subject has at least one symptom of PD.

34. A kit, array or panel for screening for a neurological disorder in subject, the kit, array or panel comprising reagents for assaying a nucleic acid sample from a subject for the presence of a genetic variation encoded by any one of SEQ ID Nos 1 -197 or 806- 916 or sub-regions thereof as set forth in Figure 9A or Figure 9D; wherein the genetic variation disrupts or modulates a genomic sequence encoded by SEQ ID Nos 198 to 805 or 908 to 1042, or an RNA transcript or polypeptide expressed therefrom.

35. A method of preventing, inhibiting or treating a subject for a movement disorder such as Parkinson's Disease, comprising: providing nucleic acid sequence information from a subject at risk of, or having or suspected of having, the movement disorder including nucleic acid sequence information on one or more genes or regions in Figure 8A or Figure 8D; identifying in the subject at least one genetic variation in the one or more genes or regions; selecting an agent or a combination of agents that modulates the one or more genes or regions; and administering the selected agent or the selected combination of agents in an amount effective to prevent, slow the progression of, or treat one or more symptoms of the movement disorder in the subject.

36. The method of claim 35 wherein the subject is heterozygous for at least one genetic variation in at least one gene that is associated with altered ubiquinone oxidoreductase activity or function, associated with lysozyme activity or function or associated with lysosomal metabolic activity or function.

37. The method of claim 35 wherein the at least one genetic variation is in one or more genes or regions associated with LYG1, L YG2, HTR7, SUMF1, UNC13C, COMMD1, HIRA, PTPRT, SGCZ, CLTCL 1, SLC25A 1, ANKRD1 1, c5orf46, JAKMIP2, SCGB3A2, SPINK4, MY01B, IQGAP2, DLGAP 1 or DLGAP5.

38. The method of claim 35 wherein the at least one genetic variation is in one or more genes or regions associated with SUMF1, GNS, ARSB, GALNS, or PSAP. 39. The method of claim 35 wherein the at least one genetic variation is in one or more genes or regions associated with NQ01 or LYG.

40. The method of any one of claims 35 to 39 wherein the at least one genetic variation comprises one or more point mutations, polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, CNVs, or loss of heterozygosity, or any combination thereof.

41. The method of any one of claims 35 to 40 wherein the agent or combination of agents includes an anti-oxidant, whey, a B vitamin, a carotene, a chloroacetic acid or a salt thereof, a dicarboxylic acid or a salt thereof, a vitamin K, a nucleoside, or a mineral:

42. The method of claim 41 wherein the B vitamin includes vitamin B1 , vitamin B2, vitamin B3, vitamin B6, vitamin B7, vitamin B9 or vitamin B12. 43. The method of claim 41 wherein the anti-oxidant includes vitamin C, vitamin B, vitamin E, a co-factor, lipoic acid, carnitine, or an organosulfur compound.

44. The method of any one of claims 35 to 43 wherein the subject is further administered a dopamine agonist or a monoamine oxidase B inhibitor.

45. The method of any one of claims 35 to 44 wherein the subject is further administered an antibody, a genetic sequence, or a combination of genetic sequences.

46. The method of any one of claims 35 to 45 wherein the selected combination of agents comprises at least two anti-oxidants and a creatine compound.

47. The method of claim 46 wherein one anti-oxidant is a co-factor, vitamin C or vitamin E. 48. The method of claim 46 wherein one anti-oxidant is an organosulfur compound.

49. A method of preventing, inhibiting or treating a symptom of a neurological disease in a subject, comprising: providing nucleic acid sequence information from a subject at risk of, or having or suspected of having, a neurological disease including nucleic acid sequence information on one or more genes or regions in Figure 8B or Figure 8C; identifying in the subject at least one genetic variation in the one or more genes or regions; and administering one or a combination of agents that includes a co- factor, whey, a B vitamin, a carotene, a chloroacetic acid or a salt thereof, a dicarboxylic acid or a salt thereof, a vitamin K, a nucleoside, a mineral, Butcher's broom (Ruscus aculeatus), curcumin, luteolin, epigallocatechin-3-gallate (antioxidant in white and green tea); thiamine, alcohol, low/no protein diets, protein supplements such as MSUD Express (manufactured by Vitaflo) that are free from branched chain amino acids leucine, isoleucine and valine, L-carnitine, salvianolic acid B, Momordica charantia (bitter melon), nicotinamide riboside, taurine, vitamin D, acetyl-L-carnitine, harpagoside, biotin, carnosine, aspirin, supplements containing antioxidants, vitamins, and other compounds such as B vitamins (B1 , B2, B3, B5, B6, and/or B9), vitamin C, vitamin E, vitamin K, acetyl L-carnitine, alpha-lipoic acid, arginine, biotin, coenzyme Q10 (e.g., in the form of ubiquinol), creatine, glycerophosphocholine, glycine, nicotine adenine dinucleotide (NADH), omega-3 fatty acids (e.g., DHA), phosphatidylserine; high fat (ketogenic) diet, or inosine supplementation, in an amount effective to prevent, jnhibit or treat at least one symptom of the neurological disease in the the subject.

50. The method of claim 49 wherein the co-factor comprises coenzyme Q10.

51. The method of claim 49 wherein the mineral comprises magnesium, selenium, calcium or phosphorus.

52. The method of claim 49 wherein the B vitamin comprises B1 , or B2 or biotin.

53. The method of claim 49 wherein the organosulfur compound comprises lipoic aicd.

54. The method of claim 49 wherein the carotene comprises beta-carotene.

55. The method of claim 49 wherein the salt of the chloroacetic acid comprises sodium dichloroacetate or potassium dichloroacetate.

56. The method of claim 49 wherein the salt of the dicarboxylic acid is sodium succinate.

57. The method of claim 49 wherein the nucleoside comprises uridine.

58. The method of claim 49 wherein a combinantion of said agents is administered.

59. The method of claim 35 wherein the at least one genetic variation is in one or more genes or regions associated with ACE2, ACY3, ALDH2, ANKRD11, ARSB, BCL2LI, CA5B, CACNA 1C, CALCRLJntergenic, CBR3, COMMD1, COX4I2, CSAD, CYP2R1, FXN, GALNS, GFRA3, GLRA3, GPR39, GRIA3, GRM1, GSTP1, HLCS, HTR7, KCNN2, L YG 1, LYG2, NDUFAF2, NDUFC2-KCTD14, NDUFS4, NLRP3, NQ01, PDE3B, PIK3C3, PRCP, PRKAA 1, PRKAG2, PSAP, S100A 12, S100A7, S100A8, S100A9, SLC13A5, SMEK2, SMPD4, SRD5A2, STEAP1, SUMF1, SUPT3H, or TXN1P.

60. A method of screening a subject for a genetic variation that disrupts or modulates one or more genes in any of Figure 7, Figure 11 D or Figure 12, comprising: assaying a nucleic acid sample obtained from the subject for a genetic variation in one or more genes in Figure 7, Figure 11 D or Figure 12, wherein the genetic variation is associated with a decreased susceptibility to developing a neurological or movement disorder or symtpoms associated with the disorder.

61. The method of claim 60 wherein the gene comprises CITED4, CTPS1 or MSRI.

Description:
METHODS AND COMPOSITIONS FOR INHIBITING AND TREATING NEUROLOGICAL

CONDITIONS [0001] The present application claims the benefit of the filing date of U.S.

application Serial No. 62/070,798, filed on Sepemtebr 5, 2014, the disclosure of which is incorporated by reference herein.

[0002] Three copies of the sequence listing (Copy 1 , Copy 2 and Copy 3) and a computer readable form (CRF copy) of the sequence listing, all in ASCII and all on CD-Rs, each containing a text file named "3885003WO1.txt", which is 656,769,024 bytes

(measured in MS-WINDOWS) and created on September 3, 2015 are incorporated herein by reference.

BACKGROUND

[0003] Genetic risk can be conferred by subtle differences in individual genomes within a population. Genes can differ between individuals due to genomic variability, the most frequent of which are due to single nucleotide polymorphisms (SNPs). SNPs can be located, on average, every 500-1000 base pairs in the human genome. Additional genetic polymorphisms in a human genome can be caused by duplication, insertion, deletion, translocation and/or inversion, of short and/or long stretches of DNA. Thus, in general, genetic variability among individuals occurs on many scales, ranging from single nucleotide changes, to gross changes in chromosome structure and function. Recently, many copy number variations (CNVs) of DNA segments, including deletions, insertions, duplications and complex multi-site variants, ranging in length from kilobases to megabases in size, have been discovered (Redon et al., Nature, 444:444 (2006) and Estivill, X. & Armengol, L. PLoS Genetics 3(10): e190 (2007)). To date, known CNVs account for over 15% of the assembled human genome (Estivill and Armengol, PLoS Genetics. 3(10): e190 (2007)). However, a majority of these variants are extremely rare and cover a small percentage of a human genome of any particular individual.

[0004] Parkinson's Disease (also known as Parkinson disease, Parkinson's, idiopathic parkinsonism, primary parkinsonism, PD, or paralysis agitans) is a degenerative disorder of the central nervous system. Parkinson's disease (PD) can be characterized by a progressive degeneration of dopaminergic neurons in the midbrain. While PD is a complex disorder of unknown etiology, it is postulated that symptom manifestation occurs after the fraction of functional dopaminergic cells falls below a threshold of twenty percent.

Symptoms of PD can include tremor, muscular rigidity, bradykinesia, akinesia, and postural instability. A hallmark of idiopathic or sporadic Parkinson's disease can be the progressive loss of dopaminergic neurons and a depletion of dopamine, more specifically in the basal ganglia, and is thought to result from a combination of genetic predisposition (Vaughn et al., Ann. Hum. Genet., 65:111 (2001 ), and environmental factors (Shapira, Adv. Neurol., 86:155 (2001 )). Thus, research efforts have focused on discovering means to prevent, protect and restore the dopaminergic cell network (Latchman et al., Rev. Neurosci., 12:69 (2001 )). As genetic polymorphisms conferring risk neurological diseases, including PD, are uncovered, genetic testing can play a role for clinical therapeutics.

[0005] There is a need to identify new treatments for neurological diseases, such as PD, and the identification of genetic risk factors that can assist in the stratification of patients for development of potential therapeutics.

SUMMARY OF THE INVENTION

[0006] As described herein, CNV analysis revealed the presence of copy number variation (CNV) in individuals with Parkinson's disease (PD), essential tremor (ET) and PD as essential tremor (ET). For instance, CNV subregions impacting 3 or more Parkinson's disease cases that were not found or occurred at a lower frequency in non-Parkinson's disease cases were identified. Also, CNV subregions impacting 1 or 2 Parkinson's disease cases that were not found in non-Parkinson's disease cases, and which subregions were associated with Parkinson's disease relevant biology (biology including apoptosis, autophagy, cell signaling (e.g., NOS, Ras or Wnt), dopaminergic function, lysosomal pathways, mitochondrial dysfunction, oxidative stress, neuroinflammation, neuroprotective factors, neurotransmitter receptors, ion channels, or ubiquitin/proteasome pathways), as well as CNV subregions occurring in intergenic regions near genes with Parkinson's disease relevant biology, were identified. As also described herein, a subset of individuals with Parkinson's disease and specific genetic variations, e.g., genetic variations in genes or regions associated with lysosomal storage or metabolism or mitochondrial dysfunction, may benefit from treatment with agents that alter lysosomal metabolism or alter mitochondrial dysfunction, e.g., complex I dysfunction. Thus, LYG1, LYG2, SUMF1, GNS/RASSF3, ARSB, GALNS, PSAP, and NUBPL genetic variations, e.g., LYG1, LYG2, SUMF1, GNS/RASSF3, ARSB, GALNS, PSAP, and NUBPL CNVs or others described herein, may be used to stratify PD patients and match certain patients with specific genetic variation(s) to certain treatments selected to modulate disease in patients with those variations. In one embodiment, a subset of individuals with Parkinson's disease and specific genetic variations in genes or regions near genes such as ACE2, ACY3, ALDH2, ANKRD11, ARSB, BCKDHB, BCL2L 1, CA5B, CACNA 1C, CALCRLJntergenic, CBR3, COMMD1, COMMD10, COX4I2, CSAD, CYP2R1, FXN, GALNS, GNSJntergenic, GFRA3, GLRA3, GPR39, GRIA3, GRM1, GSTP1, HLCS, HTR7, KCNN2, LYG1, LYG2, NDUFAF2, NDUFC2- KCTD14, NDUFS4, NLRP3, NQ01, NUBPL, PDE3B, PIK3C3, FRCP, PRKAA 1, PRKAG2, PSAP, S100A12, S100A7, S100A7A, S100A7L2, S100A8, S100A9, SLC13A5, SMEK2, SMPD4, SRD5A2, STEAP1, STEAP2, SUMF1, SUPT3H, or TXNIP, may benefit from treatment with certain agents as described herein. In one embodiment, a subset of individuals with Parkinson's disease and specific genetic variations in genes or regions near genes such as CACNA 1B, CACNA2D1 , CENPE, CERK, CFH, EPHA3, F7, GADL 1, GRIN2A, GRM5, IKBKB, KCNMA 1, KCNN3, KLRC1, LRP1, MAS1, NFKB1, NRG1, PRAME, PRTN3, PTPRC, ROCK1, RORA, TAAR1, TACR3, or USP14, may benefit from treatment with certain agents as described herein. For example, a subset of individuals with Parkinson's disease and specific genetic variations in genes or regions near genes such as CACNA 1B, CACNA2D1, CENPE, CERK, CFH, EPHA3, F7, GADL 1, GRIN2A, GRM5, IKBKB, KCNMA 1, KCNN3, KLRC1, LRP1, MAS1, NFKB1, NRG1, PRAME, PRTN3, PTPRC, ROCK1, RORA, TAAR1, TACR3, or USP14, may benefit from treatment with Butcher's broom (Ruscus aculeatus), curcumin, luteolin, epigallocatechin-3-gallate

(antioxidant in white and green tea); thiamine, alcohol, low/no protein diets, protein supplements such as MSUD Express (manufactured by Vitaflo) that are free from branched chain amino acids leucine, isoleucine and valine, L-carnitine, salvianolic acid B, Momordica charantia (bitter melon), nicotinamide riboside, taurine, vitamin D, acetyl-L-carnitine, harpagoside, biotin, carnosine, aspirin, supplements containing antioxidants, vitamins, and other compounds such as B vitamins (B1 , B2, B3, B5, B6, and/or B9), vitamin C, vitamin E, vitamin K, acetyl L-carnitine, alpha-lipoic acid, arginine, biotin, coenzyme Q10 (e.g., in the form of ubiquinol), creatine, glycerophosphocholine, glycine, nicotine adenine dinucleotide (NADH), omega-3 fatty acids (e.g., DHA), phosphatidylserine; high fat (ketogenic) diet, or inosine supplementation, or two or more combinations of those agents. In one

embodiment, the patients to be treated are those with genetic variations in genes or regions near genes such as CERK, GADL1, GRM5, or KCNN3. In one embodiment, the patients to be treated are not those with genetic variations in CACNA 1B, CACNA2D1, CENPE, CERK, CFH, EPHA3, F7, GADL1, GRIN2A, GRM5, IKBKB, KCNMA1, KCNN3, KLRC1, LRP1, MAS1, NFKB1, NRG1, PRAME, PRTN3, PTPRC, ROCK1, RORA, TAAR1, TACR3, or USP14. In one embodiment, the patients to be treated are not those with genetic variations in CERK, GADL1, GRM5, or KCNN3.

[0007] The invention provides a method of screening subjects for those with altered susceptibility to developing Parkinson's disease or ET, or those at risk of developing Parkinson's disease or ET. Another embodiment provides a method of screening subjects for those with altered susceptibility to developing one or more movement disorders that include but are not limited to PD, ET or Restless Legs Syndrome (RLS), or those at risk of developing one or more movement disorders that include but are not limited to PD, ET or RLS. The method comprises assaying at least one genetic sample of one or more subjects, nucleic acid sequence information from the one or more subjects, or providing that information, for at least one genetic variation in genes or regions associated with

Parkinson's disease, e.g., gene variations associated with one or more regions, subregions or genes in Figures 8A, 9A, 10A or 11 A, or in Figures 8D, 9D, 10D or 11 D. The presence in the genetic sample of the at least one genetic variation is used to determine whether the one or more subjects have an altered susceptibility to Parkinson's disease or ET, or are at risk of Parkinson's disease or ET. In some embodiments, determining whether the one or more subjects are at risk of Parkinson's disease or have an altered susceptibility to Parkinson's disease includes a medical history analysis and other clinical factors, e.g., in addition to the nucleic acid sequence information. In some embodiments, at least one genetic sample is collected from blood, e.g., peripheral blood mononuclear cells (PBMC) or peripheral blood lymphocytes (PBL), saliva, urine, serum, tears, skin, tissue, or hair from at least one subject. In some embodiments, assaying the at least one genetic sample of one or more subjects includes purifying the at least one genetic sample. In some embodiments, assaying the at least one genetic sample of the one or more subjects includes amplifying at least one nucleotide or a specific region of one or more chromosomes in the at least one genetic sample. In some embodiments, assaying the at least one genetic sample of the one or more subjects includes assaying an unamplified sample for at least one nucleotide or a specific region of one or more chromosomes in the at least one genetic sample. In some embodiments, assaying the at least one genetic sample for at least one genetic variation includes a microarray analysis of the at least one sample. In some embodiments, the microarray analysis comprises a comparative genomic hybridization (CGH) array analysis.

[0008] Thus, to identify patients with genetic variations that may be amenable to particular therapies, at least one genetic sample of one or more subjects may be assayed to obtain nucleic acid sequence information or that information may be provided. Nucleic acid sequence information from one or more subjects having at least one genetic variation, e.g., variations impacting or encompassing NUBPL or other genes associated with or encoding gene products in mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism, is used to determine whether the one or more subjects may benefit from a particular treatment. In one embodiment, the specific genetic variations are in regions, subregions or genes disclosed in Figures 8A, 9A, 10A, or 11 A. In one embodiment, the specific genetic variations are in regions, subregions or genes disclosed in Figures 8B, 9B, 10B, or 11B. In one embodiment, the specific genetic variations are in regions, subregions or genes disclosed in Figures 8C, 9C, 10C, or 11 C. In one embodiment, the specific genetic variations are in regions, subregions or genes disclosed in Figures 8D, 9D, 10D, or 11 D. In some embodiments, the nucleic acid sequencing information is obtained for the whole genome or whole exome from the one or more subjects. In some embodiments, the nucleic acid sequencing information has already been obtained for the whole genome or whole exome from the one or more subjects and the nucleic acid information is obtained from in silico analysis. In other embodiments, the nucleic acid sequencing information is obtained for a selected portion of the whole genome or whole exome. In some embodiments, assaying at least one genetic sample comprises obtaining the nucleic acid sequence information. In some embodiments, obtaining the nucleic acid information is determined by one or more methods selected from the group comprising PCR, sequencing, Northern blots, or any combination thereof. In some embodiments, sequencing comprises one or more high-throughput sequencing methods, Sanger sequencing, or a combination thereof. In some embodiments, at least one genetic sample is collected from blood, e.g., peripheral blood mononuclear cells (PBMC) or peripheral blood lymphocytes (PBL), saliva, urine, serum, tears, skin, tissue, or hair from at least one subject. In some embodiments, assaying the at least one genetic sample of one or more subjects includes purifying the at least one genetic sample. In some embodiments, assaying the at least one genetic sample of the one or more subjects includes amplifying at least one nucleotide or a specific region of one or more chromosomes in the at least one genetic sample. In some embodiments, assaying the at least one genetic sample of the one or more subjects includes assaying an unamplified sample for at least one nucleotide or a specific region of one or more chromosomes in the at least one genetic sample. In some embodiments, assaying the at least one genetic sample for at least one genetic variation includes a microarray analysis of the at least one sample. In some embodiments, the microarray analysis comprises a comparative genomic hybridization (CGH) array analysis. In one embodiment, the method includes detecting a genetic variation, e.g., using a multiplex ligation-dependent probe amplification (MLPA), molecular beacon, aCGH, Invader assay, ligase chain reaction (LCR), or fluorescence in situ hybridization.

[0009] In one aspect, a method for screening for a therapeutic agent useful for preventing, inhibiting or treating at least one symptom of a neurological disease (ND) such as PD or ET is provided. The method includes identifying an agent that modulates the expression of one or more genes or regions associated with mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism, modulates the expression of one or more genes encoding expression products that are part of mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism, or modulates the function or activity of expression products of the one or more genes or regions. In some embodiments, the expression products include one or more RNA transcripts for gene products associated with modulation of mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism,. In some embodiments, the expression products include one or more proteins that modulate the function or activity of mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism. In some embodiments, the agent(s) increase the expression of one or more RNA transcripts or proteins. In some embodiments, the agent(s) decrease the expression of one or more RNA transcripts or proteins. In some embodiments, an agent identified as modulating the function or activity of a mitochondrial complex is employed in a therapy based on the presence or absence of one or more genetic variations in at least one gene or regions associated with or encoding gene products of mitochondrial complex I, II, III or IV, or , or lysosomal storage or metabolism.

[0010] In some embodiments, the at least one genetic variation comprises one or more point mutations, polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. In some embodiments, the at least one genetic variation comprises one or more point mutations, single nucleotide polymorphisms (SNPs), single nucleotide variants (SNVs), polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. In some embodiments, the at least one genetic variation includes one or more CNVs in one or more genes associated with or encoding gene products in mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism. In some embodiments, the genetic variation comprises one or more CNVs that disrupt one or more genes associated with mitochondrial complex I, II, III or IV. In some embodiments, the at least one genetic variation comprises one or more CNVs that modulate the expression or function of one or more RNA transcripts of genes with gene products, or that modulate the function or activity of gene products, associated with mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism.

[0011] In one aspect, a method of treating a subject for PD or ET is provided. The method includes administering one or more agents effective to modulate the function or activity of one or more genes or regions associated with mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism, or expression products therefrom, thereby treating PD or ET. In some embodiments, the expression products include one or more RNA transcripts of a gene product found in mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism. In one embodiment, the subject has a specific genetic variation in regions, subregions or genes disclosed in Figures 8A, 9A, 10A, or 11 A. In one embodiment, the subject has a specific genetic variation in regions, subregions or genes disclosed in Figures 8B, 9B, 10B, or 11 B. In one embodiment, the subject has a specific genetic variation in regions, subregions or genes disclosed in Figures 8C, 9C, 10C, or 11C. In one

embodiment; the subject has a specific genetic variation in regions, subregions or genes disclosed in Figures 8D, 9D, 10D, or 11 D. In some embodiments, the expression products include one or more proteins expressed from a gene or regions associated with mitochondrial complex I, II, III or IV, or lysosomal storage or metabolism. In some embodiments, the agent may be an anti-oxidant, whey, a B vitamin, a carotene, a chloroacetic acid or a salt thereof, a dicarboxylic acid or a salt thereof, a vitamin K, a nucleoside, or a mineral, or a combination thereof, which may be optionally administered before, concurrently with, or subsequently to administration of an antibody, a dopamine agonist, a monoamine oxidase B inhibitor, a genetic sequence, a combination of genetic sequences, or any combination thereof.

[0012] In one aspect, the invention provides a kit, array or panel for screening for

PD or ET in a subject. In one aspect, a kit, array or panel detecs two or more, e.g., 5 to 40, 2 to 20, 5 to 10 or 5 to 15, of the genes, regions or subregions disclosed herein. In one aspect,the kit includes at least one component for assaying a genetic sample from the subject for the presence of at least one genetic variation in one or more genes associated with mitocondrial complex I, II, III or IV, e.g., mitochondrial complex I function or activity, or lysosomal storage or metabolism.

[0013] In one aspect, the patient to be treated has, or the kit, array or panel is useful for screening for, genetic variation(s) in complex I genes including, but not limited to, those encoding subunits for NADH dehydrogenase (ubiquinone). Those genetic variations may be in one or more of nuclear or mitochondrial endoded Complex I subunits and/or assembly factors: NDUFA 1, NDUFA2, NDUFA3, NDUFA4, NDUFA4L, NDUFA4L2, NDUFA5, NDUFA6, NDUFA7, NDUFA8, NDUFA9, NDUFA 10, NDUFA 11, NDUFA 12, NDUFA 13, NDUFAB†, NDUFB1, NDUFB2, NDUFB3, NDUFB4, NDUFB5, NDUFB6, NDUFB7, NDUFB8, NDUFB9, NDUFB10, NDUFB11, NDUFC1, NDUFC2, NDUFS1, NDUFS2, NDUFS3, NDUFS4, NDUFS5, NDUFS6, NDUFS7, NDUFS8, NDUFV1, NDUFV2, NDUFV3, MT-ND1, MT-ND2, MT-ND3, MT-ND4, MT-ND4L, MT-ND5, MT-ND6, ACAD9, ECSIT, FOXRED1, NDUFAF1, NDUFAF2, NDUFAF3, NDUFAF4, NDUFAF5 (aka C20orf7), NDUFAF6 (aka C8orf38), NDUFAF7 (aka C2orf56), NUBPL, TIMMDC1 (aka C3orf1), and TMEM126B. One or more agents disclosed herein may also be employed to treat subjects with genetic variations in one or subunits or assembly factors of NADH Dehydrogenase (Ubiquinone).

[0014] In one aspect, the patient to be treated has, or the kit, array or panel is useful for screening for, genetic variation(s) in genes associated with occurrence of 3- Methylglutaconic aciduria, including, but not limited to, one or more genes encoding: ATP12, ATP5E, ATPAF2, AUH, BCKDHB, CLPB, DNAJC19, OPA3, POLG, RYR1, SERAC1, SUCLA2, TAZ and TMEM70.

[0015] In one aspect, a method for screening for a therapeutic agent useful for preventing, inhibiting or treating at least one symptom of a neurological disease (ND) such as PD or ET is provided. The method includes identifying an agent that modulates the expression of one or more genes or regions associated with lysosomal storage or metabolism, modulates the expression of one or more genes encoding expression products that are part of lysosomal storage or metabolism, or modulates the function or activity of expression products of the one or more genes or regions. In some embodiments, the expression products include one or more RNA transcripts for gene products associated with modulation of lysosomal storage or metabolism. In some embodiments, the expression products include one or more proteins that modulate the function or activity of lysosomal storage or metabolism. In some embodiments, the agent(s) increase the expression of one or more RNA transcripts or proteins. In some embodiments, the agent(s) decrease the expression of one or more RNA transcripts or proteins. In some embodiments, an agent identified as modulating the function or activity of a mitochondrial complex is employed in a therapy based on the presence or absence of one or more genetic variations in at least one gene or regions associated with or encoding gene products of lysosomal storage or metabolism.

[0016] In some embodiments, the at least one genetic variation comprises one or more point mutations, polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. In some embodiments, the at least one genetic variation comprises one or more point mutations, single nucleotide polymorphisms (SNPs), single nucleotide variants (SNVs), polymorphisms, translocations, insertions, deletions, amplifications, inversions, microsatellites, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. In some embodiments, the at least one genetic variation includes one or more CNVs in one or more genes associated with or encoding gene products in lysosomal storage or metabolism. In some

embodiments, the genetic variation comprises one or more CNVs that disrupt one or more genes associated with lysosomal storage or metabolism. In some embodiments, the at least one genetic variation comprises one or more CNVs that modulate the expression or function of one or more RNA transcripts of genes with gene products, or that modulate the function or activity of gene products, associated with lysosomal storage or metabolism.

[0017] In one aspect, a method of treating a subject for PD or ET is provided. The method includes administering one or more agents effective to modulate the function or activity of one or more genes or regions associated with lysosomal storage or metabolism, or expression products therefrom, thereby treating PD or ET. In some embodiments, the expression products include one or more RNA transcripts of a gene product found in lysosomal storage or metabolism. In some embodiments, the expression products include one or more proteins expressed from a gene or regions associated with lysosomal storage or metabolism. In some embodiments, the agent may be an anti-oxidant, whey, a B vitamin, a carotene, a chloroacetic acid or a salt thereof, a dicarboxylic acid or a salt thereof, a vitamin K, a nucleoside, or a mineral, or a combination thereof, which may be optionally administered before, concurrently with, or subsequently to administration of an antibody, a dopamine agonist, a monoamine oxidase B inhibitor, a genetic sequence, a combination of genetic sequences, or any combination thereof.

[0018] In one aspect, the patient to be treated has, or a kit, array or panel is useful for screening for, genetic variation(s) in lysosomal storage or metabolic genes including but not limited to those for LYG1, LYG2, SUMF1, GNS/RASSF3, ARSB, GALNS, or PSAP, and including other sulfatases, sulfatase modifying proteins or sulfatase substrates. In one aspect, the patient to be treated has, or a kit, array or panel is useful for screening for, genetic variation(s) in one or more of ARSB, CERK, GALNS, GNS, PSAP, SCARB1, SCARB2, SMPD4, or SUMF1, including any combination thereof, and optionally one or more of GBA, GLA, GUSB, HGSNAT, IDS, IDUA, NAGLU, . or SGSH including any combination thereof. For example, substrates for SUMF1 include GALNS, ARSA, STS and ARSE, paralogs including SUMF2. Other sulfatase genes besides ARSB, GNS, and

GALNS include ARSG, ARSI, IDS, SULF1, and SULF2. Other lysozyme genes, besides LYG1 and LYG2, include LYZ, LALBA, LYZL 1, LYZL2, LYZL4, LYZL6, SPACA3, SPACA5, or SPACA5B, or other gene regions encoding enzymes that hydrolyze 1 ,4-beta linkages, e.g., between N-acetyl-D-glucosamine and N-acetylmuranic acid. In one aspect, the patient to be treated has, or a kit, array or panel is useful for screening for, genetic variation(s) in one or more potassium channel genes: KCNA7, KCND2, KCNE1, KCNIP4, KCNJ15, KCNMA 1, KCNN2, KCNN3, KCNQ5, KCNRG, or KCNS3 including any combination thereof.

[0019] In one aspect, the invention provides a kit, array or panel for screening for PD in a subject. In one aspect, the kit includes at least one component for assaying a genetic sample from the subject for the presence of at least one genetic variation in one or more genes associated with mitocondrial complex I, II, III or IV, e.g., mitochondrial complex I function or activity, or lysomal storage or metabolism.

[0020] Agents useful to treat disorders associated with a genetic variation in any of the above genes or regions may include one or more of the following agents: an antioxidant, whey, a B vitamin, a carotene, a chloroacetic acid or a salt thereof, a dicarboxylic acid or a salt thereof, a vitamin K, a nucleoside, or a mineral, or a combination thereof. For example, a subject may be administered an effective amount of riboflavin (B2), e.g., 100 to about 400 mg/day, thiamine (B1 ), e.g., about 50 to about 100 mg/day,other B vitamins (e.g., nicotinamide (B3), e.g.,. about 50 to about 100 mg/day, B6, B12 and folic acid (B9), e.g., 1 to about 10 mg/day, biotin, e.g., 2.5 to 10 mg/day ,CoQ10, for instance, about 5 to about 15 mg/kg/day, e.g., high dose CoQ10, carnitine, acetyle-L-carnitive (about 250 to about 1000 mg/day) or levo-carnitine (about 30 mg/kg/day to about 100 mg/kg/day), creatine monohydrate, lipoic acid, e.g., about 60 to about 200 mg/day up to three times per day, dichloroacetate, dimethylglycine, a whey based supplement, antioxidants like Vitamin C (ascorbic acid) or other citrates, e.g., about 100 to about 500 mg/day 1 to 3 times per day, vitamin K3, and Vitamin E (tocopherol), e.g., about 100 to about 400 lU/day 1 to 3 times per day, minerals, including but not limited to selenium, calcium, or magnesium, beta carotene, phosphorus, succinate, creatine, or uridine. In one embodiment, a subject is administered a combination of coenzyme Q10, creatine monohydrate, and lipoic acid.

[0021] Another embodiment of the invention provides a method of screening for variants that are protective against disease. That is, the aim is to identify variants that are absent or present at substantially lower frequency in affected individuals, compared to normal individuals that have a given variant at higher frequency. Such variants are likely 'protective', in that the presence of the variant in an individual appears to lessen the likelihood of a disease, rather than increase it. Such variants are often relatively common in normal populations. While causal significance for variants can be inferred even in cases where only 1 -2 affected individuals in a cohort carry the variant (if the variant results in loss of function of a gene/region for which substantial biological evidence exists that is relevant to the disease under investigation), the reverse is not true for protective variants. The presence of a variant in 1 -2 normal individuals but 0 affected individuals cannot be assumed to be protective, because the absence in the affected cohort may be the result of chance. However, for the protective variants described herein, the frequency difference between normal and affected individuals is significant enough to suggest a protective effect. A further difference exists between causal and protective variants. Causal variants within in a gene are often not identical (i.e., there can be many different mutations within the gene that can result in disease), since any that result in loss of function are likely to result in the same overall effect (loss of gene function, resulting in a phenotype). While there may be some instances wherein protective variants within a given gene are heterogeneous (i.e., multiple protective variants in different sites within or near the gene), the nature of the discovery methodology only makes it possible to identify protective variants on the basis of appreciable frequencies (e.g., 0.5-5%) in normal individuals. In the case of CNVs, those that are present at higher frequency tend to be identical in different individuals. This also has the added benefit of allowing for rapid analysis of protective variants, using an assay that screens for a gain or loss on the basis of an identical set of CNV breakpoints, in large sample numbers (cases and controls). It can be appreciated by those skilled in the art that subjects harboring a protective variant (e.g., one that results lower neuroinflammation or oxidative stress) may have a decreased susceptibility to developing PD or ET. Another embodiment provides a method of screening subjects for those with altered susceptibility to developing one or more movement disorders that include but are not limited to PD, ET or Restless Legs Syndrome (RLS), or those at risk of developing one or more movement disorders that include but are not limited to PD, ET or RLS. The method comprises assaying at least one genetic sample of one or more subjects, nucleic acid sequence information from the one or more subjects, or providing that information, for at least one genetic variation in genes or regions associated with lower risk of PD, e.g., gene variations associated with one or more regions, subregions or genes in Figures 8A, 9A, 10A or 11 A. The presence in the genetic sample of the at least one genetic variation is used to determine whether the one or more subjects have an altered susceptibility to PD or ET, or are at lower risk of PD or ET.

[0022] . Genes harboring protective variants may be different from the one or more genes that are known to cause or contribute to a given disorders, but those skilled in the art appreciate that variants within the same gene can be causal for or protective from the same disorder, or for different disorders. For example, A precedent for both causal and protective variants in a neurodegenerative disease gene is the finding of a protective variant in the Alzheimer's gene APP (Jonsson et al. Nature. 2012 Aug 2;488(7409):96-9). Protective variants also provide an opportunity to develop therapies that may treat a greater percentage of individuals diagnosed with a given disorder, such as the SLC30A8 loss-of- function mutations that were found to be protective from developing Type 2 diabetes (Flannick et al. Nat Genet. 2014 Apr;46(4):357-63).

BRIEF DESCRIPTION OF THE DRAWINGS

[0023] Figure 1. A chromosomal rearrangement disrupts NUBPL in a sporadic case of Parkinson's disease (PD). (A) Genome-wide CNV analysis revealed a complex chromosomal rearrangement in a female patient diagnosed with sporadic PD that consists of a 254 Kb loss and 134 Kb gain. It was found in 1 of 467 PD cases and in 0 of 1 ,005 controls. (B) Gene annotation for the rearrangement mapped in the UCSC genome browser (hg18) showing HEATR5A is disrupted and there is a complete loss of DTD2 and GPR33. (C) The NUBPL chromosomal rearrangement is found in PD and CI deficiency patients. PCR confirmation that the rearrangement detected in the sporadic PD case is identical to the one previously found in a CI deficiency patient (Calvo et al., Nat. Genet., 42:861 (2010)). (D) Sequence chromatograms of upstream and downstream PCR products that were generated using the PCR primers described in Tucker et al. (Hum utat.. 33:411 (2012)). Identical sequences were found at both breakpoints (upstream and downstream) in the PD patient (shown) as were found in the CI deficiency patient, including the LINE and ER2 repeat elements and a 15 bp insertion at the downstream breakpoint (sequence is the reverse complement of the Tucker et al. sequence). (E) Predicted alterations in splice events for NUBPL variants. NUBPL variants (OR > 2.0 or that are pathogenic CI deficiency mutations) were analyzed for predicted splice site event alterations using HSF. Total events corresponds to the total number of event types/positions impacted by a predicted splice event alteration. For example, for novel variant c.694-18A>T four types of events (splice site, enhancer site, silencer site, const./alt. site) at six positions (40 and 46-50) equals 8 total events. Alterations in splice events were predicted for each NUBPL variant by using a 10Obp window of sequence with the variant in position 51 to enable a comparison of variants relative to a fixed position. Exon (white-filled boxes) and intron (hatched boxes) regions are mapped according to the wild type sequence. The c.815-27T>C variant was previously reported as a pathogenic variant. Color-coding identifies if a splice event was created, destroyed, or both by the variant relative to the wild-type nucleotide. Numbers inside the colored boxes indicate the total number of predictions for each type of event listed to the left.

[0024] Figure 2. A 10 Kb exonic deletion detected in 1 Parkinson's disease (PD) case and 0 normal subjects (OR = 6.45, FET = 0.10).

[0025] Figure 3. Triplication detected in 3 PD cases and 1 normal subject (OR =

5.45, FET = 0.13). The 82 Kb triplication impacts two neighboring lysozyme G-like genes (LYG2 and LYG1 ).

[0026] Figure 4. Non-overlapping deletions detected in 3 PD cases and 0 normal subjects (OR = 15.1 , FET = 0.01 ). The deletions impact sulfatase modifying factor 1

(SUMF1 ), which has transcripts of varying length annotated in RefSeq (light gray bars in the gene annotation track) and Ensembl (dashed line in the gene annotation track) databases.

[0027] Figure 5. A 1 -2 Kb intergenic deletion detected in 5 PD cases and 1 normal subject (OR = 10.8, FET = 0.01 ). The deletion is located ~6 Kb away from the 3' end of the RASSF3 and GNS genes.

[0028] Figure 6. Known and novel PD genes in the lysosomal pathway.

[0029] Figure 7. Protective variant. A 33 Kb intergenic duplication was detected in

0 PD cases and 15 normal subjects (subset of 6 are shown, OR = 0.07, FET = 0.005).

[0030] Figure 8 shows exemplary regions with genetic variations that are associated with PD. For each variation, the following may be provided: chromosome, original CNV start, original CNV stop, original CNV size, CNV type, PD case ID, RefSeq gene symbol, and SEQ ID No. corresponding to that region. Figures 8A-D list all CNVs of interest, with the exception that, for each entry, the original CNV start and stop positions are noted, along with original CNV size, type (loss or gain), case ID and gene annotation (for the CNV-subregion NOT original CNV). Figure 8A provides CNVs corresponding regions associated with SEQ ID NOs:1 -197. Figures 8B-C provide CNVs corresponding to regions in SEQ ID NOs:1059-1340 and 1621 -2002, respectively. Figure 8D provides CNVs corresponding to regions in SEQ ID NOs:806-916. The final column in Figure 8A contains SEQ ID numbers for exemplary genes/CNV subregions, which also correspond to higher priority genes/CNV subregions. Thus, SEQ ID NO:1 has the highest priority, SEQ ID NO:2 has the next highest priority.

[0031] Figure 9 shows exemplary subregions with genetic variations that are associated with PD. For each variation, the following may be provided: chromosome, CNV subregion start, CNV subregion stop, CNV subregion size, CNV type, PD case ID(s), RefSeq gene symbol, exon overlap, NVE cases, PD cases, FET, OR, and category.

Figures 9A-D are similar to Figures 9A-D but there are a number of exceptions. Firstly, the CNV coordinates listed refer to the actual CNV-subregions found to be unique or significantly different between the disease and normal cohorts, as opposed to Figure 9, which lists the original CNVs. Secondly, an extra column details whether genie CNV- subregions of interest overlap an exon or not. Third and fourth, 2 extra columns detail the number of normal cases and the number of disease cases that harbor the relevant CNV- subregion. Finally, 3 columns report Fisher's 2-tailed Exact Test (FET), odds ratio (OR) and the Category under which the CNV-subregion falls wrt significance. Figure 9A provides CNV subregions corresponding to regions associated with SEQ ID NOs:1 -197. Figure 9B provides CNV subregions corresponding to SEQ ID NOs: 1059-1340. Figure 9C provides CNV subregions corresponding to SEQ ID NOs:1621 -2002, and Figure 9D provides CNV subregions corresponding to SEQ ID NOs:806-916.

[0032] Figure 10 is a summary of the characteristics of the regions associated with

PD. Figure 10A provides CNV subregions corresponding to regions associated with SEQ ID NOs:1 -197. Figure 10B provides CNV subregions corresponding to SEQ ID NOs:1059- 1340. Figure 10C provides CNV subregions corresponding to SEQ ID NOs:1621 -2002, and Figure 10D provides CNV subregions corresponding to SEQ ID NOs:806-916.

[0033] Figure 11 is a summary of transcripts in the regions associated with PD and

SEQ ID numbers therefor. Figure 11 A is a summary of transcripts associated with SEQ ID NOs:1 -197, e.g., those having SEQ ID Nos. 198-805. Figure 11 B is a summary of transcripts associated with SEQ ID NOs:1059-1349, e.g., SEQ ID Nos. 1341 -1620. Figure 11C is a summary of transcripts associated with SEQ ID NOs:1621 -2002, e.g., SEQ ID NOs 2003-2640. Figure 11D is a summary of transcripts associated with SEQ ID NOs:806-916, e.g., SEQ ID Nos:918-1042.

[0034] Figure 12. Protective variant. A 78-83 Kb deletion was detected in 0 PD cases and 9 normal subjects (8 of 9 subjects have an identical deletion and the ninth subject's deletion is substantially overlapped, OR = 0.11 , FET = 0.036).

[0035] Figure 13. Gene-specific therapeutic strategies.

[0036] Figure 14. Genes and neuropathological or PD relevant biology. DETAILED DESCRIPTION

[0037] Genetic risk can be conferred by subtle differences in individual genomes within a population. Genes can differ between individuals due to genomic variability, and the most frequent differences are due to single nucleotide polymorphisms (SNPs). SNPs can be located, on average, every 500-1000 base pairs in the human genome. Additional genetic polymorphisms in a human genome can be caused by duplication, insertion, deletion, translocation and/or inversion, of short and/or long stretches of DNA. Thus, in general, genetic variability among individuals occurs on many scales, ranging from single nucleotide changes, to gross changes in chromosome structure and function. Many copy number variations (CNVs) of DNA segments, including deletions, insertions, duplications and complex multi-site variants, ranging in length from kilobases to megabases in size, have been discovered (Redon et al., Nature, 444:444 (2006) and Estiviil & Armengol, PLoS Genetics, 3:1787 (2007)). Known CNVs account for over 15% of the assembled human genome (Estiviil & Armengol, supra). However, a majority of these variants are extremely rare and cover a small percentage of a human genome of any particular individual.

[0038] Described herein are methods of identifying variations in nucleic acids and genes associated with neurological disorders, in particular, PD and their use stratifying patients for therapy. Also described herein are methods and compositions for treating, inhibiting and/or preventing PD using a therapeutic modality. The present disclosure further encompasses methods of assessing an individual for probability of response to a therapeutic agent for PD, methods for predicting the effectiveness of a therapeutic agent for PD, and computer-implemented functions. Kits, arrays or panels for screening a sample from a subject to detect or determine a risk of or susceptibility to PD, or if the subject would benefit from a particular therapy, are also encompassed by the disclosure.

Neurological Disorders

[0039] As described herein, NDs, within the scope of the current disclosure can comprise:

Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis,

Adrenoleukodystrophy, Agenesis of the corpus callosum, Agnosia, Aicardi syndrome, Alexander disease, Alpers' disease, Alternating hemiplegia, Alzheimer's disease,

Amyotrophic lateral sclerosis (see Motor Neuron Disease), Anencephaly, Angelman syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid cysts, Arachnoiditis, Arnold- Chiari malformation, Arteriovenous malformation, Asperger's syndrome, Ataxia

Telangiectasia, Attention Deficit Hyperactivity Disorder, Autism, Auditory processing disorder, Autonomic Dysfunction, , Back Pain, Batten disease, Behcet's disease, Bell's palsy, Benign Essential Blepharospasm, Benign Focal Amyotrophy, Benign Intracranial Hypertension, Bilateral frontoparietal polymicrogyria, Binswanger's disease, Blepharospasm, Bloch-Sulzberger syndrome, Brachial plexus injury, Brain abscess, Brain damage, Brain injury, Brain tumor, Brown-Sequard syndrome, Canavan disease, Carpal tunnel syndrome (CTS), Causalgia, Central pain syndrome, Central pontine myelinolysis, Centronuclear myopathy, Cephalic disorder, Cerebral aneurysm , Cerebral arteriosclerosis, Cerebral atrophy, Cerebral gigantism, Cerebral palsy, Charcot-Marie-Tooth disease, Chiari malformation, Chorea, Chronic inflammatory demyelinating polyneuropathy (CIDP), Chronic pain, Chronic regional pain syndrome, Coffin Lowry syndrome, Coma, including Persistent Vegetative State, Congenital facial diplegia, Corticobasal degeneration, Cranial arteritis, Craniosynostosis, Creutzfeldt-Jakob disease, Cumulative trauma disorders, Cushing's syndrome, Cytomegalic inclusion body disease (CIBD), Cytomegalovirus Infection, , Dandy- Walker syndrome, Dawson disease, De Morsier's syndrome, Dejerine-Klumpke palsy, Dejerine-Sottas disease, Delayed sleep phase syndrome, Dementia, Dermatomyositis, Neurological Dyspraxia, Diabetic neuropathy, Diffuse sclerosis, Dysautonomia, Dyscalculia, Dysgraphia, Dyslexia, Dystonia, , Early infantile epileptic encephalopathy, Empty sella syndrome, Encephalitis, Encephalocele, Encephalotrigeminal angiomatosis, Encopresis, Epilepsy, Erb's palsy, Erythromelalgia, Essential tremor (ET), Fabry's disease, Fahr's syndrome, Fainting, Familial spastic paralysis, Febrile seizures, Fisher syndrome, Friedreich's ataxia, FART Syndrome, Gaucher's disease, Gerstmann's syndrome, Giant cell arteritis, Giant cell inclusion disease, Globoid cell Leukodystrophy, Gray matter heterotopia, Guillain-Barre syndrome, HTLV-1 associated myelopathy, Hallervorden-Spatz disease, Head injury, Headache, Hemifacial Spasm, Hereditary Spastic Paraplegia, Heredopathia atactica polyneuritiformis, Herpes zoster oticus, Herpes zoster, Hirayama syndrome, Holoprosencephaly, Huntington's disease, Hydranencephaly, Hydrocephalus,

Hypercortisolism, Hypoxia, Immune-Mediated encephalomyelitis, Inclusion body myositis, Incontinentia pigmenti, Infantile phytanic acid storage disease, Infantile Refsum disease, Infantile spasms, Inflammatory myopathy, Intracranial cyst, Intracranial hypertension, Joubert syndrome, Kearns-Sayre syndrome, Kennedy disease, Kinsboume syndrome, Klippel Feil syndrome, Krabbe disease, Kugelberg-Welander disease, Kuru, Lafora disease, Lambert-Eaton myasthenic syndrome, Landau-Kleffner syndrome, Lateral medullary (Wallenberg) syndrome, Learning disabilities, Leigh's disease, Lennox-Gastaut syndrome, Lesch-Nyhan syndrome, Leukodystrophy, Lewy body dementia, Lissencephaly, Locked-ln syndrome, Lou Gehrig's disease, Lumbar disc disease, Lyme disease - Neurological Sequelae, Machado-Joseph disease (Spinocerebellar ataxia type 3), Macrencephaly, Maple Syrup Urine Disease, Megalencephaly, Melkersson-Rosenthal syndrome, Menieres disease, Meningitis, Menkes disease, Metachromatic leukodystrophy, Microcephaly, Migraine, Miller Fisher syndrome, Mini-Strokes, Mitochondrial Myopathies, Mobius syndrome, Monomelic amyotrophy, Motor Neuron Disease, Motor skills disorder, Moyamoya disease, Mucopolysaccharidoses (including the subset referred to as Hurler Syndrome, Hurler-Scheie syndrome, Scheie syndrome, Hunter syndrome, Sanfilippo syndromes A-D, Morquio syndromes A and B, Maroteaus-Lamy syndrome, Sly syndrome, and Natowicz syndrome), Multi-lnfarct Dementia, Multifocal motor neuropathy, Multiple sclerosis, Multiple system atrophy with postural hypotension, Muscular dystrophy, Myalgic encephalomyelitis, Myasthenia gravis, Myelinoclastic diffuse sclerosis, Myoclonic

Encephalopathy of infants, Myoclonus, Myopathy, Myotubular myopathy, Myotonia congenita, Narcolepsy, Neurofibromatosis, Neuroleptic malignant syndrome, Neurological manifestations of AIDS, Neurological sequelae of lupus, Neuromyotonia, Neuronal ceroid lipofuscinosis, Neuronal migration disorders, Niemann-Pick disease, Non 24-hour sleep- wake syndrome, Nonverbal learning disorder, , O'Sullivan-McLeod syndrome, Occipital Neuralgia, Occult Spinal Dysraphism Sequence, Ohtahara syndrome, Olivopontocerebellar atrophy, Opsoclonus myoclonus syndrome, Optic neuritis, Orthostatic Hypotension, Overuse syndrome, Palinopsia, Paresthesia, Parkinson's disease, Paramyotonia

Congenita, Paraneoplastic diseases, Paroxysmal attacks, Parry-Romberg syndrome (also known as Rombergs Syndrome), Pelizaeus-Merzbacher disease, Periodic Paralyses, Peripheral neuropathy, Persistent Vegetative State, Pervasive NDs, Photic sneeze reflex, Phytanic Acid Storage disease, Pick's disease, Pinched Nerve, Pituitary Tumors, PMG, Polio, Polymicrogyria, Polymyositis, Porencephaly, Post-Polio syndrome, Postherpetic Neuralgia (PHN), Postinfectious Encephalomyelitis, Postural Hypotension, Prader-Willi syndrome, Primary Lateral Sclerosis, Prion diseases, Progressive Hemifacial Atrophy also known as Rombergs_Syndrome, Progressive multifocal leukoencephalopathy, Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Pseudotumor cerebri, Ramsay-Hunt syndrome (Type I and Type II), Rasmussen's encephalitis, Reflex sympathetic dystrophy syndrome, Refsum disease, Repetitive motion disorders, Repetitive stress injury, Restless legs syndrome, Retrovirus-associated myelopathy, Rett syndrome, Reye's syndrome, Rombergs_Syndrome, Rabies, Saint Vitus dance, Sandhoff disease, Schytsophrenia, Schilder's disease, Schizencephaly, Sensory Integration Dysfunction, Septo-optic dysplasia, Shaken baby syndrome, Shingles, Shy-Drager syndrome, Sjogren's syndrome, Sleep apnea, Sleeping sickness, Snatiation, Sotos syndrome, Spasticity, Spina bifida, Spinal cord injury, Spinal cord tumors, Spinal muscular atrophy, Spinal stenosis, Steele-Richardson-Olszewski syndrome, see Progressive Supranuclear Palsy,

Spinocerebellar ataxia, Stiff-person syndrome, Stroke, Sturge-Weber syndrome, Subacute sclerosing panencephalitis, Subcortical arteriosclerotic encephalopathy, Superficial siderosis, Sydenham's chorea, Syncope, Synesthesia, Syringomyelia, Tardive dyskinesia, Tay-Sachs disease, Temporal arteritis, Tethered spinal cord syndrome, Thomsen disease, Thoracic outlet syndrome, Tic Douloureux, Todd's paralysis, Tourette syndrome, Transient ischemic attack, Transmissible spongiform encephalopathies, Transverse myelitis, Traumatic brain injury, Tremor, Trigeminal neuralgia, Tropical spastic paraparesis, Trypanosomiasis, Tuberous sclerosis, Vasculitis including temporal arteritis, Von Hippel- Lindau disease (VHL), Viliuisk Encephalomyelitis (VE), Wallenberg's syndrome, Werdnig- Hoffman disease, West syndrome, Whiplash, Williams syndrome, Wilson's disease, X- Linked Spinal and Bulbar Muscular Atrophy, and Zellweger syndrome. In some embodiments, neurological conditions can comprise movement disorders. In one embodiment, movement disorders comprise Parkinson's Disease (PD).

[0040] The term Parkinsonism is used for a motor syndrome whose main symptoms are tremor at rest, stiffness, slowing of movement and postural instability.

Parkinsonian syndromes can be divided into four subtypes according to their origin: primary or idiopathic, secondary or acquired, hereditary parkinsonism, and parkinson plus syndromes or multiple system degeneration. Parkinson's disease is the most common form of Parkinsonism and is usually defined as "primary" Parkinsonism, meaning Parkinsonism with no external identifiable cause. As much as this can go against the definition of

Parkinson's disease as an idiopathic illness, genetic Parkinsonism disorders with a similar clinical course to PD are generally included under the Parkinson's disease label. The terms "familial Parkinson's disease" and "sporadic Parkinson's disease" can be used to differentiate genetic from truly idiopathic forms of the disease.

[0041] PD is usually classified as a movement disorder, although it also gives rise to several non-motor types of symptoms such as sensory deficits, cognitive difficulties or sleep problems. Parkinson plus diseases are primary parkinsonisms which present additional features. They include multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration and dementia with Lewy bodies.

[0042] In terms of pathophysiology, PD is considered a synucleinopathy due to an abnormal accumulation of alpha-synuclein protein in the brain in the form of Lewy bodies, as opposed to other diseases such as Alzheimer's disease where the brain accumulates tau protein in the form of neurofibrillary tangles. Nevertheless, there is clinical and pathological overlap between tauopathies and synucleinopathies. The most typical symptom of Alzheimer's disease, dementia, occurs in advanced stages of PD, while it is common to find neurofibrillary tangles in brains affected by PD.

[0043] Dementia with Lewy bodies (DLB) is another synucleinopathy that has similarities with PD, and especially with the subset of PD cases with dementia. However the relationship between PD and DLB is complex and still has to be clarified. They may represent parts of a continuum or they may be separate diseases.

[0044] Parkinson's disease affects movement, producing motor symptoms. Non- motor symptoms, which include autonomic dysfunction, neuropsychiatric problems (mood, cognition, behavior or thought alterations), and sensory and sleep difficulties, are also common.

[0045] Four motor symptoms are considered cardinal in PD: tremor, rigidity, slowness of movement, and postural instability. Tremor is the most apparent and well- known symptom. It is the most common; though around 30% of individuals with PD do not have tremor at disease onset, most develop it as the disease progresses. It is usually a rest tremor: maximal when the limb is at rest and disappearing with voluntary movement and sleep. It affects to a greater extent the most distal part of the limb and at onset typically appears in only a single arm or leg, becoming bilateral later. A feature of tremor is "pill- rolling", a term used to describe the tendency of the index finger of the hand to get into contact with the thumb and perform together a circular movement. The term derives from the similarity between the movement in PD patients and the earlier pharmaceutical technique of manually making pills.

[0046] Bradykinesia (slowness of movement) is another characteristic feature of PD, and is associated with difficulties along the whole course of the movement process, from planning to initiation and finally execution of a movement. Performance of sequential and simultaneous movement is hindered. Bradykinesia is the most disabling symptom in the early stages of the disease. Initial manifestations are problems when performing daily tasks which use fine motor control such as writing, sewing or getting dressed. Clinical evaluation is based in similar tasks such as alternating movements between both hands or both feet. Bradykinesia is not equal for all movements or times. It is modified by the activity or emotional state of the subject, to the point that some patients are barely able to walk yet can still ride a bicycle. Generally patients have less difficulty when some sort of external cue is provided.

[0047] Rigidity is stiffness and resistance to limb movement caused by increased muscle tone, an excessive and continuous contraction of muscles. In Parkinsonism the rigidity can be uniform (lead-pipe rigidity) or ratchety (cogwheel rigidity). The combination of tremor and increased tone is considered to be at the origin of cogwheel rigidity. Rigidity may be associated with joint pain; such pain being a frequent initial manifestation of the disease. In early stages of Parkinson's disease, rigidity is often asymmetrical and it tends to affect the neck and shoulder muscles prior to the muscles of the face and extremities. With the progression of the disease, rigidity typically affects the whole body and reduces the ability to move.

[0048] Postural instability is typical in the late stages of the disease, leading to impaired balance and frequent falls, and secondarily to bone fractures. Instability is often absent in the initial stages, especially in younger people. Up to 40% of the patients may experience falls and around 10% may have falls weekly, with number of falls being related to the severity of PD.

[0049] Other recognized motor signs and symptoms include gait and posture disturbances such as festination (rapid shuffling steps and a forward-flexed posture when walking), speech and swallowing disturbances including voice disorders, mask-like face expression or small handwriting, although the range of possible motor problems that can appear is large.

[0050] Parkinson's disease can cause neuropsychiatric disturbances which can range from mild to severe. This includes disorders of speech, cognition, mood, behavior, and thought. Cognitive disturbances can occur in the initial stages of the disease and sometimes prior to diagnosis, and increase in prevalence with duration of the disease. The most common cognitive deficit in affected individuals is executive dysfunction, which can include problems with planning, cognitive flexibility, abstract thinking, rule acquisition, initiating appropriate actions and inhibiting inappropriate actions, and selecting relevant sensory information. Fluctuations in attention and slowed cognitive speed are among other cognitive difficulties. Memory is affected, specifically in recalling learned information.

Nevertheless, improvement appears when recall is aided by cues. Visuospatial difficulties are also part of the disease, seen for example when the individual is asked to perform tests of facial recognition and perception of the orientation of drawn lines.

[0051] A person with PD has two to six times the risk of suffering dementia compared to the general population. The prevalence of dementia increases with duration of the disease. Dementia is associated with a reduced quality of life in people with PD and their caregivers, increased mortality, and a higher probability of needing nursing home care. Behavior and mood alterations are more common in PD without cognitive impairment than in the general population, and are usually present in PD with dementia. The most frequent mood difficulties are depression, apathy and anxiety. Impulse control behaviors such as medication overuse and craving, binge eating, hypersexuality, or pathological gambling can appear in PD and have been related to the medications used to manage the disease. Psychotic symptoms— hallucinations or delusions— occur in 4% of patients, and it is assumed that the main precipitant of psychotic phenomena in Parkinson's disease is dopaminergic excess secondary to treatment; it therefore becomes more common with increasing age and levodopa intake.

[0052] In addition to cognitive and motor symptoms, PD can impair other body functions. Sleep problems are a feature of the disease and can be worsened by medications. Symptoms can manifest in daytime drowsiness, disturbances in REM sleep, or insomnia. Alterations in the autonomic nervous system can lead to orthostatic hypotension (low blood pressure upon standing), oily skin and excessive sweating, urinary incontinence and altered sexual function. Constipation and gastric dysmotility can be severe enough to cause discomfort and even endanger health. PD is related to several eye and vision abnormalities such as decreased blink rate, dry eyes, deficient ocular pursuit (eye tracking) and saccadic movements (fast automatic movements of both eyes in the same direction), difficulties in directing gaze upward, and blurred or double vision. Changes in perception may include an impaired sense of smell, sensation of pain and paresthesia (skin tingling and numbness). All of these symptoms can occur years before diagnosis of the disease.

[0053] The primary symptoms of Parkinson's disease result from greatly reduced activity of dopamine-secreting cells caused by cell death in the pars compacta region of the substantia nigra. There are five major pathways in the brain connecting other brain areas with the basal ganglia. These are known as the motor, oculo-motor, associative, limbic and orbitofrontal circuits, with names indicating the main projection area of each circuit. All of them are affected in PD, and their disruption explains many of the symptoms of the disease since these circuits are involved in a wide variety of functions including movement, attention and learning.

[0054] Most people with Parkinson's disease have idiopathic (also termed sporadic) Parkinson's disease (having no specific known cause). A small proportion of cases, however, can be attributed to known genetic factors. Mutations in specific genes have been conclusively shown to cause PD. These genes code for alpha-synuclein (SNCA, also known as PARK1 and PARK4), parkinson protein 2 (PARK2, but also known as parkin, PRKN, as well as E3 ubiquitin ligase), leucine-rich repeat kinase 2 (LRRK2, also known as dardarin), PTEN-induced putative kinase 1 (PINK1 , also known as PARK6), parkinson protein 7 (PARK7, also known as DJ-1 ) and ATPase type 13A2 (ATP13A2), in which some mutations are referred to as Kufor-Rakeb syndrome. In most cases, people with these mutations can develop PD. With the exception of LRRK2, however, they account for only a small minority of cases of PD. The most extensively studied PD-related genes are SNCA and LRRK2. Mutations in genes including SNCA, LRRK2 and glucocerebrosidase (GBA) have been found to be risk factors for sporadic PD. Mutations in GBA are known to cause Gaucher's disease.

[0055] PD invariably progresses with time. The Hoehn and Yahr scale, which defines five stages of progression, is commonly used to estimate the progress of the disease. Motor symptoms, if not treated, advance aggressively in the early stages of the disease and more slowly later. Untreated, subjects are expected to lose independent ambulation after an average of eight years and be bedridden after ten years. However, it is uncommon to find untreated subjects nowadays. Medication has improved the prognosis of motor symptoms, while at the same time it is a new source of disability because of the undesired effects of levodopa after years of use. In subjects taking levodopa, the progression time of symptoms to a stage of high dependency from caregivers may be over 15 years. However, it is hard to predict what course the disease can take for a given subject. Age is the best predictor of disease progression. The rate of motor decline is greater in those with less impairment at the time of diagnosis, while cognitive impairment is more frequent in those who are over 70 years of age at symptom onset.

[0056] Since current therapies improve motor symptoms, disability at present is mainly related to non-motor features of the disease. Nevertheless, the relationship between disease progression and disability is not linear. Disability is initially related to motor symptoms. As the disease advances, disability is more related to motor symptoms that do not respond adequately to medication, such as swallowing/speech difficulties, and gait/balance problems; and also to motor complications, which appear in up to 50% of subjects after 5 years of levodopa usage. Finally, after ten years most subjects with the disease have autonomic disturbances, sleep problems, mood alterations and cognitive decline. All of these symptoms, especially cognitive decline, greatly increase disability. Genetic Variations Associated with Parkinson's Disease

[0057] Genomic sequences within populations exhibit variability between individuals at many locations in the genome. For example, the human genome exhibits sequence variations, which occur on average every 500 base pairs. Such genetic variations in nucleic acid sequences are commonly referred to as polymorphisms or polymorphic sites. In some embodiments, these genetic variations can be found to be associated with PD using the methods disclosed herein. In some embodiments, these genetic variations comprise point mutations, e.g., single nucleotide polymorphisms (SNPs) or single nucleotide variants (SNVs), polymorphisms, translocations, insertions, deletions, amplifications, inversions, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. In some embodiments, polymorphisms (e.g., polymorphic markers, genetic variations, or genetic variants) can comprise any nucleotide position at which two or more sequences are possible in a subject population. In some embodiments, each version of a nucleotide sequence with respect to the polymorphism can represent a specific allele of the polymorphism. In some embodiments, genomic DNA from a subject can contain two alleles for any given polymorphic marker, representative of each copy of the marker on each chromosome. In some embodiments, an allele can be a nucleotide sequence of a given location on a chromosome. Polymorphisms can comprise any number of specific alleles. In some embodiments of the disclosure, a polymorphism can be characterized by the presence of two or more alleles in a population. In some embodiments, the polymorphism can be characterized by the presence of three or more alleles. In some embodiments, the polymorphism can be characterized by four or more alleles, five or more alleles, six or more alleles, seven or more alleles, nine or more alleles, or ten or more alleles. In some embodiments an allele can be associated with one or more diseases or disorders. In some embodiments, genetic variations and alleles can be used to associate an inherited phenotype, tor example, susceptibility PD, with a responsible genotype. In some embodiments, an allele, e.g., a risk allele, can be a variant allele that is statistically associated with PD, a risk of developing PD, or an increase susceptibility to PD. In some embodiments, genetic variations can be of any measurable frequency in the population, for example, a frequency higher than 10%, a frequency between 5-10%, a frequency between 1 -5%, or frequency below 1 %. As used herein, variant alleles can be alleles that differ from a reference allele. As used herein, a variant can be a segment of DNA that differs from the reference DNA, such as a genetic variation. In some embodiments, genetic variations can be used to track the inheritance of a gene that has not yet been identified, but whose approximate location is known.

[0058] As used herein, a haplotype can be information regarding the presence or absence of one or more genetic markers in a given chromosomal region in a subject. In some embodiments, a haplotype can be a segment of DNA characterized by one or more alleles arranged along the segment, for example, a haplotype can comprise one member of the pair of alleles for each genetic variation or locus. In some embodiments, the haplotype can comprise two or more alleles, three or more alleles, four or more alleles, five or more alleles, or any combination thereof, wherein, each allele can comprise one or more genetic variations along the segment.

[0059] In some embodiments, a genetic variation can be a functional aberration that can alter gene function, gene expression, protein expression, protein function, or any combination thereof. In some embodiments, a genetic variation can be a loss-of-function mutation, gain-of-function mutation, dominant negative mutation, or reversion. In some embodiments, a genetic variation can be part of a gene's coding region or regulatory regions. Regulatory regions can control gene expression and thus protein expression. In some embodiments, a regulatory region can be a segment of DNA wherein regulatory proteins, for example, transcription factors, can bind. In some embodiments a regulatory region can be positioned near the gene being regulated, for example, positions upstream of the gene being regulated. In some embodiments, a regulatory region (e.g., enhancer element) can be several thousands of base pairs upstream or downstream of a gene.

[0060] In some embodiments, variants can include changes that affect a polypeptide or protein, such as a change in expression level, sequence, function, localization, binding partners, or any combination thereof. In some embodiments, a genetic variation can be a frameshift mutation, nonsense mutation, missense mutation, neutral mutation, or silent mutation. For example, sequence differences, when compared to a reference nucleotide sequence, can include the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of a reading frame;

duplication of all or a part of a sequence; transposition; or a rearrangement of a nucleotide sequence. Such sequence changes can alter the polypeptide encoded by the nucleic acid, for example, if the change in the nucleic acid sequence causes a frame shift, the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide. In some embodiments, a genetic variation associated with PD can be a synonymous change in one or more nucleotides, for example, a change that does not result in a change in the amino acid sequence. Such a polymorphism can, for example, alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of an encoded polypeptide. In some embodiments, a synonymous mutation can result in the protein product having an altered structure due to rare codon usage that impacts protein folding during translation, which in some cases may alter its function and/or drug binding properties if it is a drug target. In some embodiments, the changes that can alter DNA increase the possibility that structural changes, such as amplifications or deletions, occur at the somatic level. A polypeptide encoded by the reference nucleotide sequence can be a reference polypeptide with a particular reference amino acid sequence, and polypeptides encoded by variant nucleotide sequences can be variant polypeptides with variant amino acid sequences.

[0061] In some embodiments, one or more variant polypeptides or proteins can be associated with PD. In some embodiments, variant polypeptides and changes in expression, localization, and interaction partners thereof, can be used to associate an inherited phenotype, PD, with a responsible genotype. In some embodiments, an PD associated variant polypeptide can be statistically associated with a diagnosis, prognosis, or theranosis of PD.

[0062] The most common sequence variants comprise base variations at a single base position in the genome, and such sequence variants, or polymorphisms, are commonly called single nucleotide polymorphisms (SNPs) or single nucleotide variants (SNVs). In some embodiments, a SNP represents a genetic variant present at greater than or equal to 1 % occurrence in a population and in some embodiments a SNP can represent a genetic variant present at any frequency level in a population. A SNP can be a nucleotide sequence variation occurring when a single nucleotide at a location in the genome differs between members of a species or between paired chromosomes in a subject. SNPs can include variants of a single nucleotide, for example, at a given nucleotide position, some subjects can have a 'G', while others can have a 'C. SNPs can occur in a single mutational event, and therefore there can be two possible alleles possible at each SNP site; the original allele and the mutated allele. SNPs that are found to have two different bases in a single nucleotide position are referred to as biallelic SNPs, those with three are referred to as triallelic, and those with all four bases represented in the population are quadallelic. In some embodiments, SNPs can be considered neutral. In some embodiments SNPs can affect susceptibility to PD. SNP polymorphisms can have two alleles, for example, a subject can be homozygous for one allele of the polymorphism wherein both chromosomal copies of the individual have the same nucleotide at the SNP location, or a subject can be heterozygous wherein the two sister chromosomes of the subject contain different nucleotides. The SNP nomenclature as reported herein is be the official Reference SNP (rs) ID identification tag as assigned to each unique SNP by the National Center for Biotechnological Information (NCBI).

[0063] Another genetic variation of the disclosure can be copy number variations/variants (CNVs). CNVs can be alterations of the DNA of a genome that results in an abnormal number of copies of one or more sections of DNA. CNVs can be inherited or caused by de novo mutation and can be responsible for a substantial amount of human phenotypic variability, behavioral traits, and disease susceptibility. In one embodiment, CNVs of the current disclosure can be associated with risk of or susceptibility to PD. In some embodiments, CNVs can impact a single gene or include a contiguous set of genes. In some embodiments, CNVs can be caused by structural rearrangements of the genome, for example, translocations, insertions, deletions, amplifications, inversions, and interstitial deletions. In some embodiments, these structural rearrangements occur on one or more chromosomes. Low copy repeats (LCRs), which are region-specific repeat sequences, can be susceptible to these structural rearrangements, resulting in CNVs. Factors such as size, orientation, percentage similarity and the distance between the copies can influence the susceptibility of LCRs to mediate genomic rearrangement.

[0064] CNVs can account for genetic variation affecting a substantial proportion of the human genome, for example, known CNVs can cover over 15% of the human genome sequence (Estivill and Armengol, supra). CNVs can affect gene expression, phenotypic variation and adaptation by disrupting a gene or altering gene dosage, and can cause disease, for example, microdeletion and microduplication disorders, and can confer susceptibility to diseases and disorders. Updated information about the location, type, and size of known CNVs can be found in one or more databases, for example, the Database of Genomic Variants (projects.tcag.ca/variation/), which currently contains data for over 100,000 CNVs.

[0065] Other types of sequence variants can be found in the human genome and can be associated with a disease or disorder, including but not limited to, microsatellites. icrosatellite markers are stable, polymorphic, easily analyzed, and can occur regularly throughout the genome, making them especially suitable for genetic analysis. A polymorphic microsatellite can comprise multiple small repeats of bases, for example, CA repeats, at a particular site wherein the number of repeat lengths varies in a population. In some embodiments, microsatellites, for example, variable number of tandem repeats (VNTRs), can be short segments of DNA that have one or more repeated sequences, for example, about 2 to 5 nucleotides long, that can occur in non-coding DNA. In some embodiments, changes in microsatellites can occur during genetic recombination of sexual reproduction, increasing or decreasing the number of repeats found at an allele, or changing allele length.

Subjects

[0066] A subject, as used herein, can be an individual of any age from whom a sample containing nucleotides is obtained for analysis, e.g., by one or more methods described herein, so as to obtain genetic data, for example, a subject adult, child, newborn, or fetus. In some embodiments, a subject can be any target of therapeutic administration. In some embodiments, a subject can be a test subject or a reference subject. In some embodiments, a subject can be associated with PD, asymptomatic or symptomatic, have increased or decreased susceptibility to PD, be associated or unassociated with a treatment or treatment regimen, or any combination thereof. As used in the present disclosure a cohort can represent an ethnic group, a patient group, a particular age group, a group not associated with PD, a group associated with PD, a group of asymptomatic subject subjects, a group of symptomatic subject subjects, or a group or subgroup of subject subjects associated with a particular response to a treatment regimen or clinical trial. In some embodiments, a patient can be a subject afflicted with PD. In some embodiments, a patient can be a subject not afflicted with PD. In some embodiments, a subject subject can be a test subject subject, a subject patient or a subject candidate for a therapeutic, wherein genomic DNA from the subject subject, subject patient, or subject candidate is obtained for analysis by one or more methods of the present disclosure herein, so as to obtain genetic variation information of the subject, patient or candidate.

[0067] In some embodiments, the sample can be obtained prenatally from a subject fetus or embryo or from the mother, for example, from fetal or embryonic cells in the maternal circulation. In some embodiments, the sample can be obtained with the assistance of a health care provider, for example, to draw blood. In some embodiments, the sample can be obtained without the assistance of a health care provider, for example, where the sample is obtained non-invasively, such as a saliva sample, or a sample comprising buccal cells that is obtained using a buccal swab or brush, or a mouthwash sample.

[0068] The present disclosure also provides methods for assessing genetic variations in subjects who are members of a target population. Such a target population is in some embodiments a population or group of subjects at risk of developing PD, based on, for example, other genetic factors, biomarkers, biophysical parameters, family history of PD, previous screening or medical history, or any combination thereof.

[0069] In some embodiments, subjects can be from specific age subgroups, such as those over the age of 1 , over the age of 2, over the age of 3, over the age of 4, over the age of 5, over the age of 6, over the age of 7, over the age of 8, over the age of 9, over the age of 10, over the age of 15, over the age of 20, over the age of 25, over the age of 30, over the age of 35, over the age of 40, over the age of 45, over the age of 50, over the age of 55, over the age of 60, over the age of 65, over the age of 70, over the age of 75, over the age of 80, or over the age of 85. Other embodiments of the disclosure pertain to other age groups, such as subjects aged less than 85, such as less than age 80, less than age 75, less than age 70, less than age 65, less than age 60, less than age 55, less than age 50, less than age 45, less than age 40, less than age 35, less than age 30, less than age 25, less than age 20, less than age 15, less than age 10, less than age 9, less than age 8, less than age 6, less than age 5, less than age 4, less than age 3, less than age 2, or less than age 1 . Other embodiments relate to subjects with age at onset of the disease in any of particular age or age ranges defined by the numerical values described in the above or other numerical values bridging these numbers. It is also contemplated that a range of ages can be relevant in certain embodiments, such as age at onset at more than age 15 but less than age 20. Other age ranges are however also contemplated, including all age ranges bracketed by the age values listed in the above.

[0070] The genetic variations of the present disclosure found to be associated with

PD can show similar association in other subject populations. Particular embodiments comprising subject populations are thus also contemplated and within the scope of the disclosure. Such embodiments relate to subject subjects that are from one or more human populations including, but not limited to, Caucasian, European, American, Ashkenazi Jewish, Sephardi Jewish/ Eurasian, Asian, Central/South Asian, East Asian, Middle Eastern, African, Hispanic, and Oceanic populations. European populations include, but are not limited to, Swedish, Norwegian, Finnish, Russian, Danish, Icelandic, Irish, Kelt, English, Scottish, Dutch, Belgian, French, German, Spanish, Portuguese, Italian, Polish, Bulgarian, Slavic, Serbian, Bosnian, Czech, Greek and Turkish populations. The racial contribution in subject subjects can also be determined by genetic analysis, for example, genetic analysis of ancestry can be carried out using unlinked microsatellite markers such as those set out in Smith et al. (Am. J. Hum. Genet., 74:1001 (2004)).

[0071] It is also well known to the person skilled in the art that certain genetic variations have different population frequencies in different populations, or are polymorphic in one population but not in another. A person skilled in the art can however apply the methods available and as thought herein to practice the present disclosure in any given human population. This can include assessment of genetic variations of the present disclosure, so as to identify those markers that give strongest association within the specific population. Thus, the at-risk variants of the present disclosure can reside on different haplotype background and in different frequencies in various human populations.

Samples

[0072] Samples that are suitable for use in the methods described herein can be from a subject and can contain genetic or proteinaceous material, for example, genomic DNA (gDNA). Genetic material can be extracted from one or more biological samples including but not limited to, blood, saliva, urine, mucosal scrapings of the lining of the mouth, expectorant, serum, tears, skin, tissue, or hair.

[0073] In some embodiments, the sample can comprise cells or tissue, for example, cell lines. Exemplary cell types from which genetic material can be obtained using the methods described herein and include but are not limited to, a blood cell; such as a B lymphocyte, T lymphocyte, leukocyte, erythrocyte, macrophage, or neutrophil; a muscle cell such as a skeletal cell, smooth muscle cell or cardiac muscle cell; a germ cell, such as a sperm or egg; an epithelial cell; a connective tissue cell, such as an adipocyte, chondrocyte; fibroblast or osteoblast; a neuron; an astrocyte; a stromal cell; an organ specific cell, such as a kidney cell, pancreatic cell, liver cell, or a keratinocyte; a stem cell; or any cell that develops there from. A cell from which gDNA is obtained can be at a particular developmental level including, for example, a hematopoietic stem cell or a cell that arises from a hematopoietic stem cell such as a red blood cell, B lymphocyte, T lymphocyte, natural killer cell, neutrophil, basophil, eosinophil, monocyte, macrophage, or platelet. Generally any type of stem cell can be used including, without limitation, an embryonic stem cell, adult stem cell, an induced pluripotent stem cell created from an adult cell type such as fibroblasts derived from skin or pluripotent stem cell.

[0074] In some embodiments, a sample can be processed for DNA isolation, for example, DNA in a cell or tissue sample can be separated from other components of the sample. Cells can be harvested from a biological sample using standard techniques known in the art, for example, by centrifuging a cell sample and resuspending the pelleted cells, for example, in a buffered solution, for example, phosphate-buffered saline (PBS). In some embodiments, after centrifuging the cell suspension to obtain a cell pellet, the cells can be lysed to extract DNA. In some embodiments, the sample can be concentrated and/or purified to isolate DNA. All samples obtained from a subject subject, including those subjected to any sort of further processing, are considered to be obtained from the subject. In some embodiments, standard techniques and kits known in the art can be used to extract genomic DNA from a biological sample, including, for example, phenol extraction, a QIAamp® Tissue Kit (Qiagen, Chatsworth, Calif.), a Wizard® Genomic DNA purification kit (Promega), or a Qiagen Autopure method using Puregene chemistry, which can enable purification of highly stable DNA well-suited for archiving.

[0075] In some embodiments, determining the identity of an allele or determining copy number can, but need not, include obtaining a sample comprising DNA from a subject, and/or assessing the identity, copy number, presence or absence of one or more genetic variations and their chromosomal locations in the sample. The individual or organization that performs the determination need not actually carry out the physical analysis of a sample from a subject. In some embodiments, the methods can include using information obtained by analysis of the sample by a third party. In some embodiments, the methods can include steps that occur at more than one site. For example, a sample can be obtained from a subject at a first site, such as at a health care provider or at the subject's home in the case of a self-testing kit. The sample can be analyzed at the same or a second site, for example, at a laboratory or other testing facility.

Molecular Pathways in PD

[0076] The affected systems in PD include synaptic transmission, endosomal trafficking, lysosomal-autophagy, and energy metabolism or mitophagy. For example, in synaptic transmission, alpha-synuclein protein is found abundantly at the presynaptic terminals of neurons and is involved in synaptic release. At the synapse, LRRK2 levels regulate glutamate transmission, dopamine-dependent plasticity and striatal signal transduction. LRRK2 protein is also reported to interact with the dynamin superfamily of GTPases, which mediate both membrane scission in clathrin-induced endocytosis and mitochondrial fission and fusion. DNAJC6 encodes auxilin, a homolog of cyclin-G associated kinase (GAK), which is preferentially expressed in neurons and involved in clathrin uncoating and synaptic vesicle recycling. Similarly, recessively inherited mutations in SYNJ1 , encoding synaptojamin, that complexes with Hsc70 and auxilin, have been implicated in disease.

[0077] Endosomal trafficking is a highly complex and dynamic cellular process whereby vesicles or cargos that are internalized at the plasma membrane are subsequently recycled, directly or via the trans-Golgi network, and targeted for degradation by lysosomal autophagy. Neurons have a critical need to recycle membrane receptors. This can be accomplished through the clathrin-independent retromer system, a tubulovesicular tripartite complex of VPS26, VPS29 and VPS35. Multiple VPS35 subunits coalesce about FAM21 , a subunit of the WASH (Wiskott-Aldrich syndrome protein and scar homolog) complex, to mediate dynamic actin remodeling. RME-8 also binds sorting nexins and FAM21 to influence WASH and cargo trafficking. VPS35 may also physically interact with LRRK2 and Rab7L1 to influence these processes.

[0078] Lysosomes have an essential function in maintaining protein and organelle integrity within cells and impaired lysosomal function may play an important role in the pathogenesis of Parkinson's disease. The formation of intracellular aggregated alpha- synuclein or tau inclusions, albeit not a primary pathology, is also found in several ceroid lipofuscinosis disorders. These include glycolipid storage diseases such as Gaucher disease and Niemann-Pick type C that are most prevalent in Ashkenazi Jewish communities. Loss of GBA activity increases intracellular glucosylceramide accumulation, resulting in decreased lysosomal degradation and subsequent accumulation of alpha- synuclein.

[0079] Two juvenile or early-onset forms of atypical parkinsonism result directly from mutations in lysosomal proteins. X-linked parkinsonism is a consequence of splicing or protein isoform deficits in ATP6AP2 (encoding ATPase, H+ transporting, lysosomal accessory protein 2), mutations in ATP13A2 (ATPase type 13A2 gene) also result in impaired lysosomal proteolysis.

[0080] The importance of mitochondria in parkinsonism is highlighted by the identification of mutations in several genes within a common pathway for mitophagy.

Mutations in the PARK2 (parkin) gene result in a recessive form of early-onset parkinsonism. Parkin protein was first described as a proteosomal E3 ubiquitin ligase responsible for K48 substrate polyubiquination (targeting to the proteosome) and K63 monoubiquination (for signaling). PINK1 (Pteninduced kinase 1 ) and FBX07 (F-box domain-containing protein), which are also genes implicated in recessive early-on-set parkinsonism. Upstream regulators of mitophagy include TOMM7, for stabilizing PINK1 on the outer mitochondrial membrane; HSPA1 L and BAG4, which may help to regulate parkin translocation to mitochondria; and SIAH3, which is localized to mitochondria and inhibits PINK1 after mitochondrial damage.

[0081] Hexokinase activity, occurring downstream of Akt but upstream of PINK1 ,

STOML2, mitofusin1/2, GRP75, HSP60, LRPPRC, and TUFM have been nominated as downstream targets of the PINK1/parkin pathway. DJ-1 mutations, may also regulate PINK1 -dependent parkin translocation to depolarized mitochondria.

[0082] Lysosomal storage diseases (LSDs) are hereditary disorders. Most are inherited in an autosomal recessive manner. LSDs are often caused by mutations in genes encoding catabolic enzymes that are involved in degradation of macromolecules. Neuronal ceroid lipofuscinoses (NCLs)— a groups of neurodegenerative disorders that are similar to classic LSDs as they are characterized by accumulation of cellular material (namely, lipofuscin) in bodily tissues. The CNS seems to be particularly vulnerable to LSDs. LSDs are commonly caused by dysfunction in lysosomal components such as hydrolases, transporters and hydrolase activators, and lead to intralysosomal accumulation of undegraded metabolites.

[0083] Initially, classification of LSDs was made according to the nature of the accumulating storage material— as in sphingolipidoses, mucopolysaccharidoses and oligosaccharidoses. A less restrictive classification of disorders involving lysosomal storage, however, allows inclusion of diseases that display defects in cellular storage, synthetic enzymes, lysosome membrane or other membrane proteins, and trafficking. LSDs expands to include disorders that are characterized by defects in synthetic processes (such as defective GM3-synthase in GM3-gangliosidosis) or by trafficking defects

(Niemann-Pick disease, type C1 [NPC1] and NPC2), as well as including lysosomal membrane protein diseases due to faulty lysosome-associated membrane protein 1 (LAMP-1 ), LA P-2, or lysosome membrane protein II (LI P2).

[0084] It is noteworthy that variants in certain Gaucher disease associated genes were found in PD patients. PD patients or those at risk of developing PD may also have variants in genes associated with NCLs (Table 2) or human lysosome-related organelle disorders (Table 3).

retardation

[0085] In general, the NCLs are pathologically characterized by storage of autofluorescent material (including protein subunit C of mitochondrial ATP synthase or saposins) within neuronal lysosomes. 14 distinct genetic NCL variants are now recognized.

[0086] Numerous mechanisms have emerged as contributors to disease propagation, including activation of cell-death signaling, alteration of lipid content, prolonged inflammation, ER-cytosol calcium balance, and dysregulation of autophagy.

[0087] GAGs are similar to lipopolysaccharide in that they activate Toll-like receptor 4 (TLR4). Activation of this receptor leads not only to upregulation of proapoptotic ceramide in the chondrocytes, thereby causing cell death, but also to proliferation of synovial cells owing to an increase in levels of sphingosine-1 -phosphate, excess storage of heparin sulphate can lead to modulation of signaling mediated by fibroblast growth factor 2 and transforming growth factor β, which contributes to neuronal cell death, neuro- degeneration and bone pathology.

[0088] Alterations of plasma membrane lipid content and lipid raft stoichiometry can also affect receptor responses and subsequent signaling events. For instance, defects of CLN3 protein affect sphingolipid stoichiometry in lipid rafts.

[0089] Lysosphingolipids— are sphingolipids that contain a sphingoid-base free amino group. Psychosine, a lysosphingolipid that is derived from GalCer. Excessive accumulation of psychosine is associated with Krabbe disease psychosine inhibits cytokinesis via interaction with the orphan G protein-coupled receptor 8 (TDAG8).

Psychosine also inhibits protein kinase C, a signalling molecule that attenuates the response of Schwann cells and oligodendrocytes to growth factors, so excessive cellular accumulation of psychosine sensitizes these cells to apoptosis. Furthermore, through activation of phospholipase A 2 , psychosine drives an increase in arachidonic acid and lysophosphatidylcholine, which leads to caspase-3 activation, apoptosis and subsequent demyelination in both the CNS and PNS. Glucosylsphingosine accumulates in the brain and contributes to the neurodegenerative process.

[0090] The reticuloendothelial system is the major storage site in many LSDs, particularly Gaucher disease and NPA, NPB and NPC. Injury to neurons leads to activation of microglia and release of inflammatory mediators from these cells. In LSDs an increasing and lifelong storage load in neurons provides a constant stimulus for glial activation and inflammation that ultimately leads to neuronal.

[0091] In Gaucher disease, calcium release from the ER into the cytosol is increased owing to activation of the ryanodine receptor, driven by excess intracellular accumulation of glucosylceramide (GluCer). In GM1 -gangliosidosis and GM2- gangliosidosis, reduction of calcium uptake by the ER can occur owing to inhibition of the sarcoplasmic-ER calcium ATPase (SERCA) transporter. Glycosphingolipids and phospholipids can modulate ER and cytosolic calcium levels.

[0092] When taken to the extreme, autophagy can promote apoptosis either by acting alone or as an executor of programmed cell death.

[0093] Altered lipid trafficking and autophagic vacuole flux are two other mechanisms that cross paths with autophagy. Activation of autophagy is visualized as an increase in conversion of microtubule-associated protein 1 A/1 B-light chain 3 (LC3) from the cytosolic form (LC3-I) to the autophagosome-associated form (LC3-II), as well as an increase in the level of an autophagy related protein, beclinl .

Methods of Screening

[0094] As used herein, screening a subject may include diagnosing or determining, theranosing, or determining the risk of or susceptibility to developing (prognosing) PD. In particular embodiments, the disclosure is a method of determining the presence of, a risk of developing or a susceptibility to, PD, by detecting at least one genetic variation in a sample from a subject as described herein. In some embodiments, detection of particular alleles, markers, variations, or haplotypes is indicative of the presence of or susceptibility to PD.

[0095] Within any given population, there can be an absolute susceptibility of developing a disease or trait, defined as the chance of a person developing the specific disease or trait over a specified time-period. Susceptibility (e.g., being at-risk) is typically measured by looking at very large numbers of people, rather than at a particular individual. As described herein, certain copy number variations (genetic variations) are found to be useful for susceptibility assessment of PD. Susceptibility assessment can involve detecting ■ particular genetic variations in the genome of individuals undergoing assessment.

Particular genetic variations are found more frequently in individuals with PD, than in individuals without PD. Therefore, these genetic variations have predictive value for detecting EN, risk of developing PD, or a susceptibility to PD, in an individual. Without intending to be limited by theory, it is believed that the genetic variations described herein to ' be associated with susceptibility of PD represent functional variants predisposing to the disease. In some embodiments, a genetic variation can confer a susceptibility of the condition, for example, carriers of the genetic variation are at a different risk of the condition than non-carriers. In one embodiment, the presence of a genetic variation is indicative of increased susceptibility to or the presence of PD.

[0096] Screening can be performed using any method. In some embodiments, screening can be performed using Polymerase Chain Reaction (PCR). In one embodiment, screening can be performed using Array Comparative Genomic Hybridization (aCGH). In some embodiments, the genetic variation information as it relates to the current disclosure can be used in conjunction with any symptomatic screening tests.

[0097] In some embodiments, information from any of the above screening methods (e.g., specific symptoms or genetic variation data) can be used to define a subject as a test subject or reference subject. In some embodiments, information from any of the above screening methods can be used to associate a subject with a test or reference population, for example, a subject in a population.

[0098] In one embodiment, an association with PD can be determined by the statistical likelihood of the presence of a genetic variation in a subject with PD, for example, an unrelated individual or a first or second-degree relation of the subject. In some embodiments, an association with PD can be determined by determining the statistical likelihood of the absence of a genetic variation in an unaffected reference subject, for example, an unrelated individual or a first or second-degree relation of the subject. The methods described herein can include obtaining and analyzing a sample from one or more suitable reference subjects.

[0099] As used herein, susceptibility can be proneness of a subject towards the development of PD, or towards resisting development of PD, than one or more control subjects. In some embodiments, susceptibility can encompass increased susceptibility. For example, particular nucleic acid variations of the disclosure as described herein can be characteristic of increased susceptibility to development of PD. In some embodiments, susceptibility can encompass decreased susceptibility, for example, particular nucleic variations of the disclosure as described herein can be characteristic of decreased susceptibility to development of PD. As used herein, a subject at risk of developing PD has a greater chance of developing PD relative to the general population or to one or more subjects without a specific genetic variation.

[00100] As described herein, a genetic variation predictive of susceptibility to or presence of PD can be one where the particular genetic variation is more frequently present in a subject with the condition (affected), compared to the frequency of its presence in a reference group (control), such that the presence of the genetic variation is indicative of susceptibility to or presence of PD. In some embodiments, the reference group can be a population sample, for example, a random sample from the general population or a mixture of two or more samples from a population. In some embodiments, disease-free controls can be characterized by the absence of one or more specific PD-associated symptoms, for example, individuals who have not experienced symptoms associated with PD. In some embodiments, the disease-free control group is characterized by the absence of one or more PD-specific risk factors, for example, at least one genetic and/or environmental risk factor. In some embodiments, a reference sequence can be referred to for a particular site of genetic variation. In some embodiments, a reference allele can be a wild-type allele and can be chosen as either the first sequenced allele or as the allele from a control individual. In some embodiments, one or more reference subjects can be characteristically matched with one or more affected subjects, for example, with matched aged, gender or ethnicity.

[00101] A person skilled in the art can appreciate that for genetic variations with two or more alleles present in the population being studied, and wherein one allele can be found in increased frequency in a group of individuals with PD in the population, compared with ' controls, the other allele(s) of the marker can be found in decreased frequency in the group of individuals with the trait or disease, compared with controls. In such a case, one allele of the marker, for example, the allele found in increased frequency in individuals with PD, can be the at-risk allele, while the other allele(s) can be neutral or even protective.

[00102] A genetic variant associated with PD can be used to predict the susceptibility of PD for a given genotype. For any genetic variation, there can be one or more possible genotypes, for example, homozygote for the at-risk variant (e.g., in autosomal recessive disorders), heterozygote, and non-carrier of the at-risk variant. In some embodiments, susceptibility associated with variants at multiple loci can be used to estimate overall susceptibility. For multiple genetic variants, there can be k (k = 3 Λ η * 2 Λ Ρ) possible genotypes; wherein n can be the number of autosomal loci and p can be the number of gonosomal (sex chromosomal) loci. Overall susceptibility assessment calculations can assume that the relative susceptibilities of different genetic variants multiply, for example, the overall susceptibility associated with a particular genotype combination can be the product of the susceptibility values for the genotype at each locus. If the susceptibility presented is the relative susceptibility for a person, or a specific genotype for a person, compared to a reference population, then the combined susceptibility can be the product of the locus specific susceptibility values and can correspond to an overall susceptibility estimate compared with a population. If the susceptibility for a person is based on a comparison to non-carriers of the at-risk allele, then the combined susceptibility can correspond to an estimate that compares the person with a given combination of genotypes at all loci to a group of individuals who do not carry at-risk variants at any of those loci. The group of non-carriers of any at-risk variant can have the lowest estimated susceptibility and can have a combined susceptibility, compared with itself, for example, non-carriers, of 1 .0, but can have an overall susceptibility, compared with the population, of less than 1 .0.

[00103] Overall risk for multiple risk variants can be performed using standard methodology. Genetic variations described herein can form the basis of risk analysis that combines other genetic variations known to increase risk of PD, or other genetic risk variants for PD. In certain embodiments of the disclosure, a plurality of variants (genetic variations, variant alleles, and/or haplotypes) can be used for overall risk assessment. These variants are in some embodiments selected from the genetic variations as disclosed herein. Other embodiments include the use of the variants of the present disclosure in combination with other variants known to be useful for screening for PD or a susceptibility to PD. In such embodiments, the genotype status of a plurality of genetic variations, markers and/or haplotypes is determined in an individual, and the status of the individual compared with the population frequency of the associated variants, or the frequency of the variants in clinically healthy subjects, such as age-matched and sex-matched subjects.

[00104] Methods known in the art, such as the use of available algorithms and software can be used to identify, or call, significant genetic variations, including but not limited to, algorithms of DNA Analytics or DNAcopy, iPattern and/or QuantiSNP. In some embodiments, a threshold log ratio value can be used to determine losses and gains. For example, using DNA Analytics, a log2ratio cutoff of 0.25 and -0.25 to classify CNV gains and losses respectively may be used. As a further example, using DNAcopy, a log2ratio cutoff of 0.35 and -0.35 to classify CNV gains and losses respectively may be used. In some embodiments, the information and calls from two or more of the methods described herein can be compared to each other to identify significant genetic variations more or less stringently. For example, CNV calls generated by both DNA Analytics and DNAcopy algorithms may be defined as stringent CNVs. In some embodiments, significant or stringent genetic variations can be tagged as identified or called if it can be found to have a minimal reciprocal overlap to a genetic variation detected by one or more platforms and/or methods described herein. For example, a minimum of 50% reciprocal overlap can be used to tag the CNVs as identified or called.

[00105] In some embodiments, multivariate analyses or joint risk analyses, including the use of multiplicative model for overall risk assessment, and can subsequently be used to determine the overall risk conferred based on the genotype status at the multiple loci. Use of a multiplicative model, for example, assuming that the risk of individual risk variants multiply to establish the overall effect, allows for a straight-forward calculation of the overall risk for multiple markers. The multiplicative model is a parsimonious model that usually fits the data of complex traits reasonably well. Deviations from multiplicity have been rarely described in the context of common variants for common diseases, and if reported are usually only suggestive since very large sample sizes can be required to be able to demonstrate statistical interactions between loci. Assessment of risk based on such analysis can subsequently be used in the methods, uses and kits, arrays or panels of the disclosure, as described herein.

[00106] In some embodiments, the significance of increased or decreased susceptibility can be measured by a percentage. In some embodiments, a significant increased susceptibility can be measured as a relative susceptibility of at least 1.2, including but not limited to: at least 1.2, at least 1.3, at least 1.4, at least 1.5, at least 1.6, at least 1 .7, 1.8, at least 1 .9, at least 2.0, at least 2.5, at least 3.0, at least 4.0, at least 5.0, at least 6.0, at least 7.0, at least 8.0, at least 9.0, at least 10.0, and at least 15.0. In some embodiments, a relative susceptibility of at least 2.0, at least 3.0, at least 4.0, at least, 5.0, at least 6.0, or at least 10.0 is significant. Other values for significant susceptibility are also contemplated, for example, at least 2.5, 3.5, 4.5, 5.5, or any suitable other numerical values, wherein the values are also within scope of the present disclosure. In some embodiments, a significant increase in susceptibility is at least about 20%, including but not limited to about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%; 1000%, and 1500%. In one particular embodiment, a significant increase in susceptibility is at least 100%. In other embodiments, a significant increase in susceptibility is at least 200%, at least 300%, at least 400%, at least 500%, at least 700%, at least 800%, at least 900% and at least 1000%. Other cutoffs or ranges as deemed suitable by the person skilled in the art to characterize the disclosure are also contemplated, and those are also within scope of the present disclosure. In certain embodiments, a significant increase in susceptibility is characterized by a p-value, such as a p-value of less than 0.5, less than 0.4, less than 0.3, less than 0.2, less than 0.1 , less than 0.05, less than 0.01 , less than 0.001 , less than 0.0001 , less than 0.00001 , less than 0.000001 , less than 0.0000001 , less than 0.00000001 , or less than 0.000000001.

[00107] In some embodiments, an individual who is at a decreased susceptibility for or the lack of presence of PD can be an individual in whom at least one genetic variation, conferring decreased susceptibility for or the lack of presence of PD is identified. In some embodiments, the genetic variations conferring decreased susceptibility are also protective. In one aspect, the genetic variations can confer a significant decreased susceptibility of or lack of presence of PD. [0100] In some embodiments, significant decreased susceptibility can be measured as a relative susceptibility of less than 0.9, including but not limited to less than 0.9, less than 0.8, less than 0.7, less than 0,6, less than 0.5, less than 0.4, less than 0.3, less than 0.2 and less than 0.1 . In some embodiments, the decrease in susceptibility is at least 20%, including but not limited to at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% and at least 98%. Other cutoffs or ranges as deemed suitable by the person skilled in the art to characterize the disclosure are however also contemplated, and those are also within scope of the present disclosure. In certain embodiments, a significant decrease in susceptibility is characterized by a p-value, such as a p-value of less than 0.05, less than 0.01 , less than 0.001 , less than 0.0001 , less than 0.00001 , less than 0.000001 , less than 0.0000001 , less than 0.00000001 , or less than 0.000000001 . Other tests for significance can be used, for example, a Fisher's exact test. Other statistical tests of significance known to the skilled person are also contemplated and are also within scope of the disclosure.

[0101] In some embodiments, the significance of increased or decreased susceptibility can be determined according to the ratio of measurements from a test subject to a reference subject. In one embodiment, losses or gains of one or more CNVs can be determined according to a threshold log2 ratio determined by these measurements. In some embodiments, a log2 ratio value greater than 0.35 is indicative of a gain of one or more CNVs. In some embodiments, a log2 ratio value less than -0.35 is indicative of a loss of one or more CNVs.

[0102] In some embodiments, the combined or overall susceptibility associated with a plurality of variants associated with PD can also be assessed, for example, the genetic variations described herein to be associated with susceptibility to PD can be combined with other common genetic risk factors. Combined risk for such genetic variants can be estimated in an analogous fashion to the methods described herein.

[0103] Calculating risk conferred by a particular genotype for the individual can be based on comparing the genotype of the individual to previously determined risk expressed, for example, as a relative risk (RR) or an odds ratio (OR), for the genotype, for example, for a heterozygous carrier of an at-risk variant for PD. An odds ratio can be a statistical measure used as a metric of causality. For example, in genetic disease research it can be used to convey the significance of a variant in a disease cohort relative to an

unaffected/normal cohort. The calculated risk for the individual can be the relative risk for a subject, or for a specific genotype of a subject, compared to the average population. The average population risk can be expressed as a weighted average of the risks of different genotypes, using results from a reference population, and the appropriate calculations to calculate the risk of a genotype group relative to the population can then be performed. Alternatively, the risk for an individual can be based on a comparison of particular genotypes, for example, heterozygous carriers of an at-risk allele of a marker compared with non-carriers of the at-risk allele. Using the population average can, in certain embodiments, be more convenient, since it provides a measure which can be easy to interpret for the user, for example, a measure that gives the risk for the individual, based on his/her genotype, compared with the average in the population.

[0 04] ■ In certain embodiments of the disclosure, a genetic variation is correlated to

PD by referencing genetic variation data to a look-up table that comprises correlations between the genetic variation and PD. The genetic variation in certain embodiments comprises at least one indication of the genetic variation. In some embodiments, the table comprises a correlation for one genetic variation. In other embodiments, the table comprises a correlation for a plurality of genetic variations. In both scenarios, by referencing to a look-up table that gives an indication of a correlation between a genetic variation and PD, a risk for PD, or a susceptibility to PD, can be identified in the individual from whom the sample is derived.

[0105] The screening applications of PD-associated genetic variations, as described herein, can, for example, be performed by an individual, a health professional, or a third party, for example, a service provider who interprets genotype information from the subject.

[0106] A medical professional can initiate or modify treatment after receiving information regarding a subject's screening for PD, for example. In some embodiments, a medical professional can recommend a change in therapy. In some embodiments, a medical professional can enroll a subject in a clinical trial for, by way of example, detecting correlations between a haplotype as described herein and any measurable or quantifiable parameter relating to the outcome of the treatment as described above.

[0107] ' Also provided herein are databases that include a list of genetic variations as described herein, and wherein the list can be largely or entirely limited to genetic variations identified as useful for screening PD as described herein. The list can be stored, for example, on a flat file or computer-readable medium. The databases can further include information regarding one or more subjects, for example, whether a subject is affected or unaffected, clinical information such as endophenotype, age of onset of symptoms, any treatments administered and outcomes, for example, data relevant to pharmacogenomics, diagnostics, prognostics or theranostics, and other details, for example, data about the disorder in the subject, or environmental or other genetic factors. The databases can be used to detect correlations between a particular haplotype and the information regarding the subject. [0108] The methods described herein can also include the generation of reports for use, for example, by a subject, care giver, or researcher, that include information regarding a subject's genetic variations, and optionally further information such as treatments administered, treatment history, medical history, predicted response, and actual response. The reports can be recorded in a tangible medium, e.g., a computer-readable disk, a solid state memory device, or an optical storage device.

Methods of Screening using Variations in Polypeptides and/or RNA

[0109] In some embodiments of the disclosure, screening of PD can be made by examining or comparing changes in expression, localization, binding partners, and composition of a polypeptide encoded by a nucleic acid associated with PD, for example, in those instances where the genetic variations of the present disclosure results in a change in the composition or expression of the polypeptide and/or RNA, for example, mRNAs, miRNAs, and other noncoding RNAs (ncRNAs). Thus, screening of PD can be made by examining expression and/or composition of one of these polypeptides and/or RNA, or another polypeptide and/or RNA encoded by a nucleic acid associated with PD, in those instances where the genetic variation of the present disclosure results in a change in the expression, localization, binding partners, and/or composition of the polypeptide and/or RNA. In some embodiments, screening can comprise diagnosing a subject. In some embodiments, screening can comprise determining a prognosis of a subject, for example, determining the susceptibility of developing PD. In some embodiments, screening can comprise theranosing a subject.

[0110] The genetic variations described herein that show association to PD can play a role through their effect on one or more of these nearby genes. For example, while not intending to be limited by theory, it is generally expected that a deletion of a chromosomal segment comprising a particular gene, or a fragment of a gene, can either result in an altered composition or expression, or both, of the encoded protein and/or mRNA. Likewise, duplications, or high number copy number variations, are in general expected to result in increased expression of encoded polypeptide and/or RNA if the duplication encompasses the whole gene. It is also known to those skilled in the art that segments of DNA can be duplicated, triplicated, quadruplicated, or amplified many times and result in increasingly higher levels of expression of the gene if it is encompassed by these multiplicated segments of DNA. Those skilled in the art also know that one or both breakpoints of a duplication or other level of amplification can disrupt a gene and thus result in loss of function, such as the expressed protein encoded by the transcript is truncated. Further, those skilled in the art anticipate that an amplified segment of DNA can occur in tandem (e.g., multiple gene copies adjacent to each other on the chromosome) or can insert into a site far away from the original chromosomal location or even on another chromosome. Thus, in some cases a gene not contained within the amplified segment of DNA is impacted by the chromosomal rearrangement. Such complex rearrangements can be mapped, for example, by fluorescence in situ hybridization (FISH) methods. Other possible mechanisms affecting genes within or near a genetic variation region include, for example, effects on transcription, effects on RNA splicing, alterations in relative amounts of alternative splice forms of mRNA, effects on RNA stability, effects on transport from the nucleus to cytoplasm, and effects on the efficiency and accuracy of translation. Thus, DNA variations can be detected directly, using the subjects unamplified or amplified genomic DNA, or indirectly, using RNA or DNA obtained from the subject's tissue(s) that are present in an aberrant form or expression level as a result of the genetic variations of the disclosure showing association to PD.

[0111] In some embodiments, the genetic variations of the disclosure showing association to PD can affect the expression of a gene within the genetic variation region. Certain genetic variation regions can have flanking duplicated segments, and genes within such segments can have altered expression and/or composition as a result of such genomic alterations. It is also well known that regulatory elements affecting gene expression can be located far away, even as far as tens or hundreds of kilobases away, from the promoter region of a gene. Thus, regulatory elements for genes that are located outside the genetic variation region can be located within the genetic variation, and thus affect the expression of genes located outside the genetic variation. It is thus contemplated that the detection of the genetic variations described herein, can be used for assessing expression for one or more of associated genes.

[0112] In some embodiments, genetic variations of the disclosure showing association to PD can affect protein expression at the translational level. It can be appreciated by those skilled in the art that this can occur by increased or decreased expression of one or more microRNAs (miRNAs) that regulates expression of a protein known to be important, or implicated, in the cause, onset, or progression of PD. Increased or decreased expression of the one or more miRNAs can result from gain or loss of the whole miRNA gene, disruption of a portion of the gene (e.g., by an indel or CNV), or even a single base change (SNP or SNV) that produces an altered, non-functional or aberrant functioning miRNA sequence. It can also be appreciated by those skilled in the art that the expression of protein, for example, one known to cause EN by increased or decreased expression, can result due to a genetic variation that results in alteration of an existing miRNA binding site within the protein's mRNA transcript, or even creates a new miRNA binding site that leads to aberrant protein expression.

[0113] A variety of methods can be used for detecting protein composition and/or expression levels, including but not limited to enzyme linked immunosorbent assays (ELISA), Western blots, spectroscopy, mass spectrometry, peptide arrays, colorimetry, electrophoresis, isoelectric focusing, immunoprecipitations, immunoassays, and immunofluorescence and other methods well-known in the art. A test sample from a subject can be assessed for the presence of an alteration in the expression and/or an alteration in composition of the polypeptide encoded by a nucleic acid associated with PD. An "alteration" in the polypeptide expression or composition, as used herein, refers to an alteration in expression or composition in a test sample, as compared to the expression or composition of the polypeptide in a control sample. Such alteration can, for example, be an alteration in the quantitative polypeptide expression or can be an alteration in the qualitative polypeptide expression, for example, expression of a mutant polypeptide or of a different splicing variant, or a combination thereof. In some embodiments, screening for PD can be made by detecting a particular splicing variant encoded by a nucleic acid associated with PD, or a particular pattern of splicing variants.

[0114] Antibodies can be polyclonal or monoclonal and can be labeled or unlabeled. An intact antibody or a fragment thereof can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled as previously described herein. Other non-limiting examples of indirect labeling include detection of a primary antibody using a labeled secondary antibody, for example, a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin.

Detecting Genetic Variations Associated with Parkinson's Disease

[0115] Described herein, are methods that can be used to detect genetic variations. Detecting specific genetic variations, for example, polymorphic markers and/or haplotypes, copy number, absence or presence of an allele, or genotype associated with PD as described herein, can be accomplished by methods known in the art for analyzing nucleic acids and/or detecting sequences at polymorphic or genetically variable sites, for example, amplification techniques, hybridization techniques, sequencing, arrays, or any combination thereof. Thus, by use of these methods disclosed herein or other methods available to the person skilled in the art, one or more alleles at polymorphic markers, including microsatellites, SNPs, CNVs, or other types of genetic variations, can be identified in a sample obtained from a subject.

Nucleic Acids

[0116] The nucleic acids and polypeptides described herein can be used in methods and kits, arrays or panels of the present disclosure. In some embodiments, aptamers that specifically bind the nucleic acids and polypeptides described herein can be used in methods and kits, arrays or panels of the present disclosure. As used herein, a nucleic acid can comprise a deoxyribonucleotide (DNA) or ribonucleotide (RNA), whether singular or in polymers, naturally occurring or non-naturally occurring, double-stranded or single-stranded, coding, for example, a translated gene, or non-coding, for example, a regulatory region, or any fragments, derivatives, mimetics or complements thereof. In some embodiments, nucleic acids can comprise oligonucleotides, nucleotides, polynucleotides, nucleic acid sequences, genomic sequences, antisense nucleic acids, DNA regions, probes, primers, genes, regulatory regions, introns, exons, open-reading frames, binding sites, target nucleic acids and allele-specific nucleic acids.

[0117] "Isolated" nucleic acids, as used herein, are separated from nucleic acids that normally flank the gene or nucleotide sequence (as in genomic sequences) and/or has been completely or partially purified from other transcribed sequences (e.g., as in an RNA library). For example, isolated nucleic acids of the disclosure can be substantially isolated with respect to the complex cellular milieu in which it naturally occurs, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized. In some instances, the isolated material can form part of a composition, for example, a crude extract containing other substances, buffer system or reagent mix. In some embodiments, the material can be purified to essential homogeneity using methods known in the art, for example, by poly aery I amide gel electrophoresis (PAGE) or column chromatography (e.g., HPLC). With regard to genomic DNA (gDNA), the term "isolated" also can refer to nucleic acids that are separated from the chromosome with which the genomic DNA is naturally associated. For example, the isolated nucleic acid molecule can contain less than about 250 kb, 200 kb, 150 kb, 100 kb, 75 kb, 50 kb, 25 kb, 10 kb, 5 kb, 4 kb, 3 kb, 2kb, 1 kb, 0.5 kb or 0.1 kb of the nucleotides that flank the nucleic acid molecule in the gDNA of the cell from which the nucleic acid molecule is derived.

[0118] Nucleic acids can be fused to other coding or regulatory sequences can be considered isolated. For example, recombinant DNA contained in a vector is included in the definition of "isolated" as used herein. In some embodiments, isolated nucleic acids can include recombinant DNA molecules in heterologous host cells or heterologous organisms, as well as partially or substantially purified DNA molecules in solution. Isolated nucleic acids also encompass in vivo and in vitro RNA transcripts of the DNA molecules of the present disclosure. An isolated nucleic acid molecule or nucleotide sequence can be synthesized chemically or by recombinant means. Such isolated nucleotide sequences can be useful, for example, in the manufacture of the encoded polypeptide, as probes for isolating homologous sequences (e.g., from other mammalian species), for gene mapping (e.g., by in situ hybridization with chromosomes), or for detecting expression of the gene, in tissue (e.g., human tissue), such as by Northern blot analysis or other hybridization techniques disclosed herein. The disclosure also pertains to nucleic acid sequences that hybridize under high stringency hybridization conditions, such as for selective hybridization, to a nucleotide sequence described herein Such nucleic acid sequences can be detected and/or isolated by allele- or sequence-specific hybridization (e.g., under high stringency conditions). Stringency conditions and methods for nucleic acid hybridizations are well known to the skilled person (see, e.g., Current Protocols in Molecular Biology. Ausubel, F. et al., John Wiley & Sons, (1998), and Kraus, M. and Aaronson, S., Methods Enzvmol, 200:546 (1991 ), the entire teachings of which are incorporated by reference herein.

[0119] Calculations of "identity" or "percent identity" between two or more nucleotide or amino acid sequences can be determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first sequence). The nucleotides at corresponding positions are then compared, and the percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions x 100). For example, a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.

[0120] In some embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%, of the length of the reference sequence. The actual comparison of the two sequences can be accomplished by well-known methods, for example, using a mathematical algorithm. A non-limiting example of such a mathematical algorithm is described in Karlin, and Altschul, Proc. Natl. Acad. Sci. USA. 90:5873 (1993). Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0), as described in Altschul et al., Nucleic Acids Res., 25:3389 (1997). When utilizing BLAST and Gapped BLAST programs, any relevant parameters of the respective programs (e.g., NBLAST) can be used. For example, parameters for sequence comparison can be set at score = 100, word length = 12, or can be varied (e.g., W=5 or W=20). Other examples include the algorithm of Myers and Miller, CABIOS (1989), ADVANCE, ADAM, BLAT, and FASTA. In some embodiments, the percent identity between two amino acid sequences can be accomplished using, for example, the GAP program in the GCG software package (Accelrys, Cambridge, UK).

[0121] "Probes" or "primers" can be oligonucleotides that hybridize in a base- specific manner to a complementary strand of a nucleic acid molecule. Probes can include primers, which can be a single-stranded oligonucleotide probe that can act as a point of initiation of template-directed DNA synthesis using methods including but not limited to, polymerase chain reaction (PCR) and ligase chain reaction (LCR) for amplification of a target sequence. It can be appreciated by those skilled in the art that probes for detection of amplified or unamplified nucleic acid molecules can also include an Invader oligonucleotide and probe pair. Oligonucleotides, as described herein, can include segments or fragments of nucleic acid sequences, or their complements. In some embodiments, DNA segments can be between 5 and 10,000 contiguous bases, and can range from 5, 10, 12, 15, 20, or 25 nucleotides to 10, 15, 20, 25, 30, 40, 50, 100, 200, 500, 1000 or 10,000 nucleotides. In addition to DNA and RNA, probes and primers can include polypeptide nucleic acids (PNA), as described in Nielsen, P. et al., Science 254: 1497-1500 (1991 ). A probe or primer can comprise a region of nucleotide sequence that hybridizes to at least about 15, typically about 20-25, and in certain embodiments about 40, 50 or 75, consecutive nucleotides of a nucleic acid molecule.

[0122] The present disclosure also provides isolated nucleic acids, for example, probes or primers, that contain a fragment or portion that can selectively hybridize to a nucleic acid that comprises, or consists of, a nucleotide sequence, wherein the nucleotide sequence can comprise at least one polymorphism or polymorphic allele contained in the genetic variations described herein or the wild-type nucleotide that is located at the same position, or the compliments thereof. In some embodiments, the probe or primer can be at least 70% identical, at least 80% identical, at least 85% identical, at least 90% identical, or at least 95% identical, to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence.

[0123] In one embodiment, a nucleic acid probe can be an oligonucleotide capable of hybridizing with a complementary region of a gene associated with PD containing a genetic variation described herein. The nucleic acid fragments of the disclosure can be used as probes or primers in assays such as those described herein.

[0124] The nucleic acids of the disclosure, such as those described above, can be identified and isolated using standard molecular biology techniques well known to the skilled person. In some embodiments, DNA can be amplified and/or can be labeled (e.g., radiolabeled, fluorescently labeled) and used as a probe for screening, for example, a cDNA library derived from an organism. cDNA can be derived from mRNA and can be contained in a suitable vector. For example, corresponding clones can be isolated, DNA obtained fallowing in vivo excision, and the cloned insert can be sequenced in either or both orientations by art-recognized methods to identify the correct reading frame encoding a polypeptide of the appropriate molecular weight. Using these or similar methods, the polypeptide and the DNA encoding the polypeptide can be isolated, sequenced and further characterized.

[0125] In some embodiments, nucleic acid can comprise one or more

polymorphisms, variations, or mutations, for example, sjngle nucleotide polymorphisms (SNPs), copy number variations (CNVs), for example, insertions, deletions, inversions, and translocations. In some embodiments, nucleic acids can comprise analogs, for example, phosphorothioates, phosphoramidates, methyl phosphonate, chiral methyl phosphonates, 2-0-methyl ribonucleotides, or modified nucleic acids, for example, modified backbone residues or linkages, or nucleic acids combined with carbohydrates, lipids, protein or other materials, or peptide nucleic acids (PNAs), for example, chromatin, ribosomes, and transcriptosomes. In some embodiments nucleic acids can comprise nucleic acids in various structures, for example, A DNA, B DNA, Z-form DNA, siRNA, tRNA, and ribozymes. In some embodiments, the nucleic acid may be naturally or non-naturally polymorphic, for example, having one or more sequence differences, for example, additions, deletions and/or substitutions, as compared to a reference sequence. In some embodiments, a reference sequence can be based on publicly available information, for example, the U.C. Santa Cruz Human Genome Browser Gateway (genome.ucsc.edu/cgi-bin/hgGateway) or the NCBI website (www.ncbi.nlm.nih.gov). In some embodiments, a reference sequence can be determined by a practitioner of the present disclosure using methods well known in the art, for example, by sequencing a reference nucleic acid.

[0126] In some embodiments, a probe can hybridize to an allele, SNP, or CNV as described herein. In some embodiments, the probe can bind to another marker sequence associated with PD as described herein.

[0127] One of skill in the art would know how to design a probe so that sequence specific hybridization can occur only if a particular allele is present in a genomic sequence from a test sample. The disclosure can also be reduced to practice using any convenient genotyping method, including commercially available technologies and methods for genotyping particular genetic variations.

[0128] Control probes can also be used, for example, a probe that binds a less variable sequence, for example, a repetitive DNA associated with a centromere of a chromosome, can be used as a control. In some embodiments, probes can be obtained from commercial sources. In some embodiments, probes can be synthesized, for example, chemically or in vitro, or made from chromosomal or genomic DNA through standard techniques. In some embodiments sources of DNA that can be used include genomic DNA, cloned DNA sequences, somatic cell hybrids that contain one, or a part of one, human chromosome along with the normal chromosome complement of the host, and chromosomes purified by flow cytometry or microdissection. The region of interest can be isolated through cloning, or by site-specific amplification using PCR.

[0129] One or more nucleic acids for example, a probe or primer, can also be labeled, for example, by direct labeling, to comprise a detectable label. A detectable label can comprise any label capable of detection by a physical, chemical, or a biological process for example, a radioactive label, such as 32P or 3H, a fluorescent label, such as FITC, a chromophore label, an affinity-ligand label, an enzyme label, such as alkaline phosphatase, ' horseradish peroxidase, or I2 galactosidase, an enzyme cofactor label, a hapten conjugate label, such as digoxigenin or dinitrophenyl, a Raman signal generating label, a magnetic label, a spin label, an epitope label, such as the FLAG or HA epitope, a luminescent label, a heavy atom label, a nanoparticle label, an electrochemical label, a light scattering label, a spherical shell label, semiconductor nanocrystal label, such as quantum dots (described in U.S. Pat. No. 6,207,392), and probes labeled with any other signal generating label known to those of skill in the art, wherein a label can allow the probe to be visualized with or without a secondary detection molecule. A nucleotide can be directly incorporated into a probe with standard techniques, for example, nick translation, random priming, and PCR labeling.

[0130] Non-limiting examples of label moieties useful for detection include, without limitation, suitable enzymes such as horseradish peroxidase, alkaline phosphatase, beta- galactosidase, or acetylcholinesterase; members of a binding pair that are capable of forming complexes such as streptavidin/biotin, avidin/biotin or an antigen/antibody complex including, for example, rabbit IgG and anti-rabbit IgG; fluorophores such as umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, tetramethyl rhodamine, eosin, green fluorescent protein, erythrosin, coumarin, methyl coumarin, pyrene, malachite green, stilbene, lucifer yellow, Cascade Blue, Texas Red, dichlorotriazinylamine fluorescein, dansyl chloride, phycoerythrin, fluorescent lanthanide complexes such as those including

Europium and Terbium, cyanine dye family members, such as Cy3 and Cy5, molecular beacons and fluorescent derivatives thereof, as well as others known in the art as described, for example, in Principles of Fluorescence Spectroscopy, Joseph R. Lakowicz (Editor), Plenum Pub Corp, 2nd edition (July 1999) and the 6th Edition of the Molecular Probes Handbook by Richard P. Hoagland; a luminescent material such as luminol; light scattering or plasmon resonant materials such as gold or silver particles or quantum dots; or radioactive material include 14C, 1231, 1241, 1251, Tc99m, 32P, 33P, 35S or 3H.

[0131] Other labels can also be used in the methods of the present disclosure, for example, backbone labels. Backbone labels comprise nucleic acid stains that bind nucleic acids in a sequence independent manner. Non-limiting examples include intercalating dyes such as phenanthridines and acridines (e.g., ethidium bromide, propidium iodide, hexidium iodide, dihydroethidium, ethidium homodimer-1 and -2, ethidium monoazide, and ACMA) ; some minor grove binders such as indoles and imidazoles (e.g., Hoechst 33258, Hoechst 33342, Hoechst 34580 and DAPI) ; and miscellaneous nucleic acid stains such as acridine orange (also capable of intercalating), 7-AAD, actinomycin D, LDS751 , and

hydroxystilbamidine. All of the aforementioned nucleic acid stains are commercially available from suppliers such as Molecular Probes, Inc. Still other examples of nucleic acid stains include the following dyes from Molecular Probes: cyanine dyes such as SYTOX Blue, SYTOX Green, SYTOX Orange, POPO-1 , POPO-3, YOYO-1 , YOYO-3, TOTO-1 , TOTO-3, JOJO-1 , LOLO-1 , BOBO-1 , BOBO-3, PO-PRO-1 , PO-PRO-3, BO-PRO-1 , BO- PRO-3, TO-PRO-1 , TO-PRO-3, TO-PRO-5, JO-PRO-1 , LO-PRO-1 , YO-PRO-1 , YO-PRO-3, PicoGreen, OliGreen, RiboGreen, SYBR Gold, SYBR Green I, SYBR Green II, SYBR DX, SYTO-40, -41 , -42, -43, -44, -45 (blue), SYTO-13, -16, -24, -21 , -23, -12, -11, -20, -22, -15, - 14, -25 (green), SYTO-81 , -80, -82, -83, -84, -85 (orange), SYTO-64, -17, -59, -61 , -62, -60, -63 (red).

[0132] In some embodiments, fluorophores of different colors can be chosen, for example, 7-amino-4-methylcoumarin-3-acetic acid (AMCA), 5-(and-6)-carboxy-X- rhodamine, lissamine rhodamine B, 5-(and-6)-carboxyfluorescein, fluorescein-5- isothiocyanate (FITC), 7-diethylaminocoumarin-3-carboxylic acid, tetramethylrhodamine-5- (and-6)-isothiocyanate, 5-(and-6)-carboxytetramethylrhodamine, 7-hydroxycoumarin-3- carboxylic acid, 6-[fluorescein 5-(and-6)-carboxamido]hexanoic acid, N-(4,4-difluoro-5,7- dimethyl-4-bora-3a,4a diaza-3-indacenepropionic acid, eosin-5-isothiocyanate, erythrosin-5- isothiocyanate, TRITC, rhodamine, tetramethylrhodamine, R-phycoerythrin, Cy-3, Cy-5, Cy- 7, Texas Red, Phar-Red, allophycocyanin (APC), and CASCADE blue acetylazide, such that each probe in or not in a set can be distinctly visualized. In some embodiments, fluorescently labeled probes can be viewed with a fluorescence microscope and an appropriate filter for each fluorophore, or by using dual or triple band-pass filter sets to observe multiple fluorophores. In some embodiments, techniques such as flow cytometry can be used to examine the hybridization pattern of the probes.

[0133] In other embodiments, the probes can be indirectly labeled, for example, with biotin or digoxygenin, or labeled with radioactive isotopes such as 32P and/or 3H. As a non-limiting example, a probe indirectly labeled with biotin can be detected by avidin conjugated to a detectable marker. For example, avidin can be conjugated to an enzymatic marker such as alkaline phosphatase or horseradish peroxidase. In some embodiments, enzymatic markers can be detected using colorimetric reactions using a substrate and/or a catalyst for the enzyme. In some embodiments, catalysts for alkaline phosphatase can be used, for example, 5-bromo-4-chloro-3-indolylphosphate and nitro blue tetrazolium. In some embodiments, a catalyst can be used for horseradish peroxidase, for example, diaminobenzoate.

Methods of Detecting Genetic Variations

[0134] In some embodiments, standard techniques for genotyping for the presence genetic variations, for example, amplification, can be used. Amplification of nucleic acids can be accomplished using methods known in the art. Generally, sequence information from the region of interest can be used to design oligonucleotide primers that can be identical or similar in sequence to opposite strands of a template to be amplified. In some embodiments, amplification methods can include but are not limited to, fluorescence- based techniques utilizing PCR, for example, ligase chain reaction (LCR), Nested PCR, transcription amplification, self-sustained sequence replication, nucleic acid based sequence amplification (NASBA), and multiplex ligation-dependent probe amplification (MLPA). Guidelines for selecting primers for PCR amplification are well known in the art. In some embodiments, a computer program can be used to design primers, for example, Oligo (National Biosciences, Inc., Plymouth Minn.), MacVector (Kodak/IBI), and GCG suite of sequence analysis programs.

[0135] In some embodiments, commercial methodologies available for genotyping, for example, SNP genotyping, can be used, but are not limited to, TaqMan genotyping assays (Applied Biosystems), SNPlex platforms (Applied Biosystems), gel electrophoresis, capillary electrophoresis, size exclusion chromatography, mass spectrometry, for example, MassARRAY system (Sequenom), minisequencing methods, real-time Polymerase Chain Reaction (PCR), Bio-Plex system (BioRad), CEQ and SNPstream systems (Beckman), array hybridization technology, for example, Affymetrix GeneChip (Perlegen), BeadArray Technologies, for example, lllumina GoldenGate and Infinium assays, array tag technology, Multiplex Ligation-dependent Probe Amplification (MLPA), and endonuclease-based fluorescence hybridization technology (Invader; Third Wave/Hologic). PCR can be a procedure in which target nucleic acid is amplified in a manner similar to that described in U.S. Pat. No. 4,683,195 and subsequent modifications of the procedure described therein. In some embodiments, real-time quantitative PGR can be used to determine genetic variations, wherein quantitative PCR can permit both detection and quantification of a DNA sequence in a sample, for example, as an absolute number of copies or as a relative amount when normalized to DNA input or other normalizing genes. In some embodiments, methods of quantification can include the use of fluorescent dyes that can intercalate with double-stranded DNA, and modified DNA oligonucleotide probes that can fluoresce when hybridized with a complementary DNA.

[0136] In some embodiments of the disclosure, a sample containing genomic DNA obtained from the subject can be collected and PCR can used to amplify a fragment of nucleic acid that comprises one or more genetic variations that can be indicative of a susceptibility to PD. In some embodiments, detection of genetic variations can be accomplished by expression analysis, for example, by using quantitative PCR. In some embodiments, this technique can assess the presence of an alteration in the expression or composition of one or more polypeptides or splicing variants encoded by a nucleic acid associated with PD.

[0137] In one embodiment, the DNA template of a sample from a subject containing a SNP can be amplified by PCR prior to detection with a probe. In such an embodiment, the amplified DNA serves as the template for a detection probe and, in some embodiments, an enhancer probe. Certain embodiments of the detection probe, the enhancer probe, and/or the primers used for amplification of the template by PCR can comprise the use of modified bases, for example, modified A, T, C, G, and U, wherein the use of modified bases can be useful for adjusting the melting temperature of the nucleotide probe and/or primer to the template DNA. In one embodiment, modified bases are used in the design of the detection nucleotide probe. Any modified base known to the skilled person can be selected in these methods, and the selection of suitable bases is well within the scope of the skilled person based on the teachings herein and known bases available from commercial sources as known to the skilled person.

[0138] In some embodiments, identification of genetic variations can be accomplished using hybridization methods. The presence of a specific marker allele or a particular genomic segment comprising a genetic variation, or representative of a genetic variation, can be indicated by sequence-specific hybridization of a nucleic acid probe specific for the particular allele or the genetic variation in a nucleic acid containing sample that has or has not been amplified but methods described herein. The presence of more than one specific marker allele or several genetic variations can be indicated by using two or more sequence-specific nucleic acid probes, wherein each is specific for a particular allele and/or genetic variation.

[0139] Hybridization can be performed by methods well known to the person skilled in the art, for example, hybridization techniques such as fluorescent in situ hybridization (FISH), Southern analysis, Northern analysis, or in situ hybridization. In some embodiments, hybridization refers to specific hybridization, wherein hybridization can be performed with no mismatches. Specific hybridization, if present, can be using standard methods. In some embodiments, if specific hybridization occurs between a nucleic acid probe and the nucleic acid in the sample, the sample can contain a sequence that can be complementary to a nucleotide present in the nucleic acid probe. In some embodiments, if a nucleic acid probe can contain a particular allele of a polymorphic marker, or particular alleles for a plurality of markers, specific hybridization is indicative of the nucleic acid being completely complementary to the nucleic acid probe, including the particular alleles at polymorphic markers within the probe. In some embodiments a probe can contain more than one marker allele of a particular haplotype, for example, a probe can contain alleles complementary to 2, 3, 4, 5 or all of the markers that make up a particular haplotype. In some embodiments detection of one or more particular markers of the haplotype in the sample is indicative that the source of the sample has the particular haplotype.

[0140] In some embodiments, PCR conditions and primers can be developed that amplify a product only when the variant allele is present or only when the wild type allele is present, for example, allele-specific PCR. In some embodiments of allele-specific PCR, a method utilizing a detection oligonucleotide probe comprising a fluorescent moiety or group at its 3' terminus and a quencher at its 5' terminus, and an enhancer oligonucleotide, can be employed, as described by Kutyavin et al. (Nucleic Acid Res.. 34:e128 (2006)).

[0141] An allele-specific primer/probe can be an oligonucleotide that is specific for particular a polymorphism can be prepared using standard methods. In some

embodiments, allele-specific oligonucleotide probes can specifically hybridize to a nucleic acid region that contains a genetic variation. In some embodiments, hybridization conditions can be selected such that a nucleic acid probe can specifically bind to the sequence of interest, for example, the variant nucleic acid sequence.

[0142] In some embodiments, allele-specific restriction digest analysis can be used to detect the existence of a polymorphic variant of a polymorphism, if alternate polymorphic variants of the polymorphism can result in the creation or elimination of a restriction site. Allele-specific restriction digests can be performed, for example, with the particular restriction enzyme that can differentiate the alleles. In some embodiments, PCR can be used to amplify a region comprising the polymorphic site, and restriction fragment length polymorphism analysis can be conducted. In some embodiments, for sequence variants that do not alter a common restriction site, mutagenic primers can be designed that can introduce one or more restriction sites when the variant allele is present or when the wild type allele is present.

[0143] In some embodiments, fluorescence polarization template-directed dye- terminator incorporation (FP-TDI) can be used to determine which of multiple polymorphic variants of a polymorphism can be present in a subject. Unlike the use of allele-specific probes or primers, this method can employ primers that can terminate adjacent to a polymorphic site, so that extension of the primer by a single nucleotide can result in incorporation of a nucleotide complementary to the polymorphic variant at the polymorphic site.

[0144] In some embodiments, DNA containing an amplified portion can be dot- blotted, using standard methods and the blot contacted with the oligonucleotide probe. The presence of specific hybridization of the probe to the DNA can then be detected. The methods can include determining the genotype of a subject with respect to both copies of the polymorphic site present in the genome, wherein if multiple polymorphic variants exist at a site, this can be appropriately indicated by specifying which variants are present in a subject. Any of the detection means described herein can be used to determine the genotype of a subject with respect to one or both copies of the polymorphism present in the subject's genome.

[0145] In some embodiments, a peptide nucleic acid (PNA) probe can be used in addition to, or instead of, a nucleic acid probe in the methods described herein. A PNA can be a DNA mimic having a peptide-like, inorganic backbone, for example, N-(2-aminoethyl) glycine units with an organic base (A, G, C, T or U) attached to the glycine nitrogen via a methylene carbonyl linker.

[0146] Nucleic acid sequence analysis can also be used to detect genetic variations, for example, genetic variations can be detected by sequencing exons, introns, 5' untranslated sequences, or 3' untranslated sequences. One or more methods of nucleic acid analysis that are available to those skilled in the art can be used to detect genetic variations, including but not limited to, direct manual sequencing, automated fluorescent sequencing, single-stranded conformation polymorphism assays (SSCP); clamped denaturing gel electrophoresis (CDGE); denaturing gradient gel electrophoresis (DGGE), two-dimensional gel electrophoresis (2DGE or TDGE); conformational sensitive gel electrophoresis (CSGE); denaturing high performance liquid chromatography (DHPLC), infrared matrix-assisted laser desorption/ionization (IR- ALDI) mass spectrometry, mobility shift analysis, quantitative real-time PCR, restriction enzyme analysis, heteroduplex analysis; chemical mismatch cleavage (CMC), RNase protection assays, use of polypeptides that recognize nucleotide mismatches, allele-specific PCR, real-time pyrophosphate DNA sequencing, PCR amplification in combination with denaturing high performance liquid chromatography (dHPLC), and combinations of such methods.

[0147] Sequencing can be accomplished through classic Sanger sequencing methods, which are known in the art. In one embodiment sequencing can be performed using high-throughput sequencing methods some of which allow detection of a sequenced nucleotide immediately after or upon its incorporation into a growing strand, for example, detection of sequence in substantially real time or real time. In some cases, high throughput sequencing generates at least 1 ,000, at least 5,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 sequence reads per hour; with each read being at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read (or 500 - 1 ,000 bases per read for 454). [0148] High-throughput sequencing methods can include but are not limited to,

Massively Parallel Signature Sequencing (MPSS, Lynx Therapeutics), Polony sequencing, 454 pyrosequencing, lllumina (Solexa) sequencing, SOUD sequencing, on semiconductor sequencing, DNA nanoball sequencing, Helioscope™ single molecule sequencing, Single Molecule SMRT™ sequencing, Single Molecule real time (RNAP) sequencing, Nanopore DNA sequencing, and/or sequencing by hybridization, for example, a non-enzymatic method that uses a DNA microarray, or microfluidic Sanger sequencing.

[0149] In some embodiments, high-throughput sequencing can involve the use of technology available by Helicos Biosciences Corporation (Cambridge, Mass.) such as the Single Molecule Sequencing by Synthesis (SMSS) method. SMSS is unique because it allows for sequencing the entire human genome in up to 24 hours. This fast sequencing method also allows for detection of a SNP/nucleotide in a sequence in substantially real time or real time. Finally, SMSS is powerful because, like the MIP technology, it does not use a pre-amplification step prior to hybridization. SMSS does not use any amplification. SMSS is described in US Publication Application Nos. 20060024711 ; 20060024678;

20060012793; 20060012784; and 20050100932. In some embodiments, high-throughput sequencing involves the use of technology available by 454 Life Sciences, Inc. (a Roche company, Branford, Conn.) such as the PicoTiterPlate device which includes a fiber optic plate that transmits chemiluminescent signal generated by the sequencing reaction to be recorded by a CCD camera in the instrument. This use of fiber optics allows for the detection of a minimum of 20 million base pairs in 4.5 hours.

[0150] In some embodiments, PCR-amplified single-strand nucleic acid can be hybridized to a primer and incubated with a polymerase, ATP sulfurylase, luciferase, apyrase, and the substrates luciferin and adenosine 5' phosphosulfate. Next,

deoxynucleotide triphosphates corresponding to the bases A, C, G, and T (U) can be added sequentially. A base incorporation can be accompanied by release of pyrophosphate, which can be converted to ATP by sulfurylase, which can drive synthesis of oxyluciferin and the release of visible light. Since pyrophosphate release can be equimolar with the number of incorporated bases, the light given off can be proportional to the number of nucleotides adding in any one step. The process can repeat until the entire sequence can be determined. In some embodiments, pyrosequencing can be utilized to analyze amplicons to determine whether breakpoints are present. In some embodiments, pyrosequencing can map surrounding sequences as an internal quality control.

[0151] Pyrosequencing analysis methods are known in the art. Sequence analysis can include a four-color sequencing by ligation scheme (degenerate ligation), which involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes can be performed. At any given cycle, the population of nonamers that is used can be structured such that the identity of one of its positions can be correlated with the identity of the fluorophore attached to that nonamer. To the extent that the ligase discriminates for complementarily at that queried position, the fluorescent signal can allow the inference of the identity of the base. After performing the ligation and four-color imaging, the anchor primer: nonamer complexes can be stripped and a new cycle begins. Methods to image sequence information after performing ligation are known in the art.

[0152] In some embodiments, analysis by restriction enzyme digestion can be used to detect a particular genetic variation if the genetic variation results in creation or elimination of one or more restriction sites relative to a reference sequence. In some embodiments, restriction fragment length polymorphism (RFLP) analysis can be conducted, wherein the digestion pattern of the relevant DNA fragment indicates the presence or absence of the particular genetic variation in the sample.

[0153] In some embodiments, arrays of oligonucleotide probes that can be complementary to target nucleic acid sequence segments from a subject can be used to identify genetic variations. In some embodiments, an array of oligonucleotide probes comprises an oligonucleotide array, for example, a microarray. In some embodiments, the present disclosure features arrays that include a substrate having a plurality of addressable areas, and methods of using them. At least one area of the plurality includes a nucleic acid probe that binds specifically to a sequence comprising a genetic variation, and can be used to detect the absence or presence of the genetic variation, for example, one or more SNPs, microsatellites, or CNVs, as described herein, to determine or identify an allele or genotype. For example, the array can include one or more nucleic acid probes that can be used to detect a genetic variation associated with a gene and/or product of a gene listed in Figures 8-11. In some embodiments, the array can further comprise at least one area that includes a nucleic acid probe that can be used to specifically detect another marker associated with PD, as described herein.

[0154] Microarray hybridization can be performed by hybridizing a nucleic acid of interest, for example, a nucleic acid encompassing a genetic variation, with the array and detecting hybridization using nucleic acid probes. In some embodiments, the nucleic acid of interest is amplified prior to hybridization. Hybridization and detecting can be carried out according to standard methods described in Published PCT Applications: WO 92/10092 and WO 95/11995, and U.S. Pat. No. 5,424,186. For example, an array can be scanned to determine the position on the array to which the nucleic acid hybridizes. The hybridization data obtained from the scan can be, for example, in the form of fluorescence intensities as a function of location on the array. [0155] Arrays can be formed on substrates fabricated with materials such as paper; glass; plastic, for example, polypropylene, nylon, or polystyrene; polyacrylamide; nitrocellulose; silicon; optical fiber; or any other suitable solid or semisolid support; and can be configured in a planar, for example, glass plates or silicon chips); or three dimensional, for example, pins, fibers, beads, particles, microtiter wells, and capillaries, configuration.

[0156] Methods for generating arrays are known in the art and can include for example; photolithographic methods (U.S. Pat. Nos. 5,143,854, 5,510,270 and 5,527,681 ); mechanical methods, for example, directed-flow methods (U.S. Pat. No. 5,384,261 ); pin- based methods (U.S. Pat. No. 5,288,514); bead-based techniques (PCT US/93/04145); solid phase oligonucleotide synthesis methods; or by other methods known to a person skilled in the art (see, e.g., Bier et al., Adv. Biochem. Eng. Biotechnol.. 109:433 (2008); Hoheisel, Nat. Rev. Genet.. 7: 200 (2006); Fan et al., Methods Enzvmol.. 410:57 (2006); Raqoussis & Elvidge, Expert Rev. Mol. Design, 6: 145-52 (2006); Mockler et al., Genomics, 85:1 (2005), and references cited therein, the entire teachings of each of which are incorporated by reference herein). Many additional descriptions of the preparation and use of oligonucleotide arrays for detection of polymorphisms can be found, for example, in U.S. Pat. Nos. 6,858,394; 6,429,027; 5,445,934; 5,700,637; 5,744,305; 5,945,334; 6,054,270; 6,300,063; 6,733,977; 7,364,858, EP 619 321 , and EP 373 203, the entire teachings of which are incorporated by reference herein. Methods for array production, hybridization, and analysis are also described in Snijders et al., Nat. Genetics, 29:263 (2001 ); Klein et al., Proc. Natl. Acad. Sci. USA, 96:4494 (1999); Albertson et al., Breast Cancer Research and Treatment, 78:289 (2003); and Snijders et al., "BAC microarray based comparative genomic hybridization," in: Zhao et al. (eds), Bacterial Artificial Chromosomes: Methods and Protocols, Methods in Molecular Biology, Humana Press, 2002.

[0157] In some embodiments, oligonucleotide probes forming an array can be attached to a substrate by any number of techniques, including, but not limited to, in situ synthesis, for example, high-density oligonucleotide arrays, using photolithographic techniques; spotting/printing a medium to low density on glass, nylon, or nitrocellulose; by masking; and by dot-blotting on a nylon or nitrocellulose hybridization membrane. In some embodiments, oligonucleotides can be immobilized via a linker, including but not limited to, by covalent, ionic, or physical linkage. Linkers for immobilizing nucleic acids and polypeptides, including reversible or cleavable linkers, are known in the art (U.S. Pat. No. 5,451 ,683 and W098/20019). In some embodiments, oligonucleotides can be non- covalently immobilized on a substrate by hybridization to anchors, by means of magnetic beads, or in a fluid phase, for example, in wells or capillaries.

[0158] An array can comprise oligonucleotide hybridization probes capable of specifically hybridizing to different genetic variations. In some embodiments, oligonucleotide arrays can comprise a plurality of different oligonucleotide probes coupled to a surface of a substrate in different known locations. In some embodiments, oligonucleotide probes can exhibit differential or selective binding to polymorphic sites, and can be readily be designed by one of ordinary skill in the art, for example, an

oligonucleotide that is perfectly complementary to a sequence that encompasses a polymorphic site, for example, a sequence that includes the polymorphic site, within it, or at one end, can hybridize preferentially to a nucleic acid comprising that sequence, as opposed to a nucleic acid comprising an alternate polymorphic variant.

[0159] In some embodiments, arrays can include multiple detection blocks, for example, multiple groups of probes designed for detection of particular polymorphisms. In some embodiments, these arrays can be used to analyze multiple different polymorphisms. In some embodiments, detection blocks can be grouped within a single array or in multiple, separate arrays, wherein varying conditions, for example, conditions optimized for particular polymorphisms, can be used during hybridization. General descriptions of using oligonucleotide arrays for detection of polymorphisms can be found, for example, in U.S. Pat. Nos. 5,858,659 and 5,837,832. In addition to oligonucleotide arrays, cDNA arrays can be used similarly in certain embodiments.

[0160] The methods described herein can include but are not limited to providing an array as described herein; contacting the array with a sample, and detecting binding of a nucleic acid from the sample to the array. In some embodiments, the method can comprise amplifying nucleic acid from the sample, for example, a region associated with PD or a region that includes another region associated with PD. In some embodiments, the methods described herein can include using an array that can identify differential expression patterns or copy numbers of one or more genes in samples from control and affected individuals. For example, arrays of probes to a marker described herein can be used to identify genetic variations between DNA from an affected subject, and control DNA obtained from an individual that does not have PD. Since the nucleotides on the array can contain sequence tags, their positions on the array can be accurately known relative to the genomic sequence.

[0161] In some embodiments, it can be desirable to employ methods that can detect the presence of multiple genetic variations, for example, polymorphic variants at a plurality of polymorphic sites, in parallel or substantially simultaneously. In some embodiments, these methods can comprise oligonucleotide arrays and other methods, including methods in which reactions, for example, amplification and hybridization, can be performed in individual vessels, for example, within individual wells of a multi-well plate or other vessel. [0162] Determining the identity of a genetic variation can also include or consist of reviewing a subject's medical history, where the medical history includes information regarding the identity, copy number, presence or absence of one or more alleles or SNPs in the subject, e.g., results of a genetic test.

[0163] Genetic variations can also be identified using any of a number of methods well known in the art. For example, genetic variations available in public databases, which can be searched using methods and custom algorithms or algorithms known in the art, can be used. In some embodiments, a reference sequence can be from, for example, the human draft genome sequence, publicly available in various databases, or a sequence deposited in a database such as GenBank.

Methods of Detecting CNVs

[0164] Detection of genetic variations, specifically CNVs, can be accomplished by one or more suitable techniques described herein. Generally, techniques that can selectively determine whether a particular chromosomal segment is present or absent in an individual can be used for genotyping CNVs. Identification of novel copy number variations can be done by methods for assessing genomic copy number changes.

[0165] In some embodiments, methods include but are not limited to, methods that can quantitatively estimate the number of copies of a particular genomic segment, but can also include methods that indicate whether a particular segment is present in a sample or not. In some embodiments, the technique to be used can quantify the amount of segment present, for example, determining whether a DNA segment is deleted, duplicated, or triplicated in subject, for example, Fluorescent In Situ Hybridization (FISH) techniques, and other methods described herein.

[0 66] In some embodiments, other genotyping technologies can be used for detection of CNVs, including but not limited to, karyotype analysis, Molecular Inversion Probe array technology, for example, Affymetrix SNP Array 6.0, and BeadArray

Technologies, for example, lllumina GoldenGate and Infinium assays, as can other platforms such as NimbleGen HD2.1 or HD4.2, High-Definition Comparative Genomic Hybridization (CGH) arrays (Agilent Technologies), tiling array technology (Affymetrix), multiplex ligation-dependent probe amplification (MLPA), Invader assay, qPCR, or fluorescence in situ hybridization. In one embodiment, Array Comparative Genomic Hybridization (aCGH) methods can be used. As described herein, karyotype analysis can be a method to determine the content and structure of chromosomes in a sample. In some embodiments, karyotyping can be used, in lieu of aCGH, to detect translocations, which can be copy number neutral (balanced translocations), and therefore, not detectable by aCGH. Information about amplitude of particular probes, which can be representative of particular alleles, can provide quantitative dosage information for the particular allele, and by consequence, dosage information about the CNV in question, since the marker can be selected as a marker representative of the CNV and can be located within the CNV. In some embodiments, if the CNV is a deletion, the absence of particular marker allele is representative of the deletion. In some embodiments, if the CNV is a duplication or a higher order copy number variation, the signal intensity representative of the allele correlating with the CNV can represent the copy number. A summary of methodologies commonly used is provided in Perkel (J. Nature Methods. 5:447 (2008)).

[0167] PCR assays can be utilized to detect CNVs and can provide an alternative to array analysis. In particular, PCR assays can enable detection of precise boundaries of gene/chromosome variants, at the molecular level, and which boundaries are identical in different individuals. PCR assays can be based on the amplification of a junction fragment present only in individuals that carry a deletion. This assay can convert the detection of a loss by array CGH to one of a gain by PCR.

[0168] Examples of PCR techniques that can be used in the present disclosure include, but are not limited to quantitative PCR, real-time quantitative PCR (qPCR), quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, PCR-RFLP/RT-PCR-RFLP, hot start PCR and Nested PCR. Other suitable amplification methods include the ligase chain reaction (LCR), ligation mediated PCR (LM-PCR), degenerate oligonucleotide probe PCR (DOP-PCR), transcription amplification, self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP- PCR), arbitrarily primed polymerase chain reaction (AP-PCR) and nucleic acid based sequence amplification (NABSA).

[0169] . Alternative methods for the simultaneous interrogation of multiple regions include quantitative multiplex PCR of short fluorescent fragments (QMPSF), multiplex amplifiable probe hybridization (MAPH) and multiplex ligation-dependent probe

amplification (MLPA), in which copy-number differences for up to 40 regions can be scored in one experiment. Another approach can be to specifically target regions that harbor known segmental duplications, which are often sites of copy-number variation. By targeting the variable nucleotides between two copies of a segmental duplication (called paralogous sequence variants) using a SNP-genotyping method that provides independent fluorescence intensities for the two alleles, it is possible to detect an increase in intensity of one allele compared with the other.

[0170] In some embodiments, the amplified piece of DNA can be bound to beads using the sequencing element of the nucleic acid tag under conditions that favor a single amplified piece of DNA molecule to bind a different bead and amplification occurs on each bead. In some embodiments, such amplification can occur by PCR. Each bead can be placed in a separate well, which can be a picoliter-sized well. In some embodiments, each bead is captured within a droplet of a PCR-reaction-mixture-in-oil-emulsion and PCR amplification occurs within each droplet. The amplification on the bead results in each bead carrying at least one million, at least 5 million, or at least 10 million copies of the single amplified piece of DNA molecule.

[0171] In some embodiments where PCR occurs in oil-emulsion mixtures, the emulsion droplets are broken, the DNA is denatured and the beads carrying single-stranded nucleic acids clones are deposited into a well, such as a picoliter-sized well, for further analysis according to the methods described herein. These amplification methods allow for the analysis of genomic DNA regions. Methods for using bead amplification followed by fiber optics detection are described in Margulies et al., Nature. 15:437(7057) :376-80 (2005), and as well as in US Publication Application Nos. 20020012930; 20030068629;

20030100102; 20030148344; 20040248161 ; 20050079510, 20050124022; and

20060078909.

[0172] Another variation on the array-based approach can be to use the hybridization signal intensities that are obtained from the oligonucleotides employed on Affymetrix SNP arrays or in lllumina Bead Arrays. Here hybridization intensities are compared with average values that are derived from controls, such that deviations from these averages indicate a change in copy number. As well as providing information about copy number, SNP arrays have the added advantage of providing genotype information. For example, they can reveal loss of heterozygosity, which could provide supporting evidence for the presence of a deletion, or might indicate segmental uniparental disomy (which can recapitulate the effects of structural variation in some genomic regions - Prader- Willi and Angelman syndromes, for example).

[0173] Many of the basic procedures followed in microarray-based genome profiling are similar, if not identical, to those followed in expression profiling and SNP analysis, including the use of specialized microarray equipment and data-analysis tools. Since microarray-based expression profiling has been well established in the last decade, much can be learned from the technical advances made in this area. Examples of the use of microarrays in nucleic acid analysis that can be used are described in U.S. Pat. No.

6,300,063, U.S. Pat. No. 5,837,832, U.S. Pat. No. 6,969,589, U.S. Pat. No. 6,040,138, U.S. Pat. No. 6,858,412, U.S. application Ser. No. 08/529,115, U.S. application Ser. No.

10/272,384, U.S. application Ser. No. 10/045,575, U.S. application Ser. No. 10/264,571 and U.S. application Ser. No. 10/264,574. It should be noted that there are also distinct differences such as target and probe complexity, stability of DNA over RNA, the presence of repetitive DNA and the need to identify single copy number alterations in genome profiling. [0174] In one embodiment, the genetic variations detected comprise CNVs and may be detected using array CGH. In some embodiments, array CGH can be been implemented using a wide variety of techniques. The initial approaches used arrays produced from large-insert genomic clones such as bacterial artificial chromosomes (BACs). Producing sufficient BAC DNA of adequate purity to make arrays is arduous, so several techniques to amplify small amounts of starting material have been employed. These techniques include ligation-mediated PCR (Snijders et al, Nat. Genet.. 29:263-64),- degenerate primer PCR using one or several sets of primers, and rolling circle amplification. BAC arrays that provide complete genome tiling paths are also available. Arrays made from less complex nucleic acids such as cDNAs, selected PCR products, and

oligonucleotides can also be used. Although most CGH procedures employ hybridization with total genomic DNA, it is possible to use reduced complexity representations of the genome produced by PCR techniques. Computational analysis of the genome sequence can be used to design array elements complementary to the sequences contained in the representation. Various SNP genotyping platforms, some of which use reduced complexity genomic representations, can be useful for their ability to determine both DNA copy number and allelic content across the genome. In some embodiments, small amounts of genomic DNA can be amplified with a variety of whole genome amplification methods prior to CGH analysis of the sample.

[0175] The different basic approaches to array CGH provide different levels of performance, so some are more suitable for particular applications than others. The factors that determine performance include the magnitudes of the copy number changes, their genomic extents, the state and composition of the specimen, how much material is available for analysis, and how the results of the analysis can be used. Many applications use reliable detection of copy number changes of much less than 50%, a higher stringency than for other microarray technologies. Note that technical details are extremely important and different implementations of methods using the same array CGH approach can yield different levels of performance. Various CGH methods are known in the art and are equally applicable to one or more methods of the present disclosure For example, CGH methods are disclosed in U.S. Pat. Nos. 7,957,913, 7,910,353, 7,238,484, 7,702,468, 7,034,144;

7,030,231 ; 7,011 ,949; 7,014,997; 6,977,148; 6,951 ,761 ; and 6,916,621 , the disclosure from each of which is incorporated by reference herein in its entirety.

[0176] The data provided by array CGH are quantitative measures of DNA sequence dosage. Array CGH provides high-resolution estimates of copy number aberrations, and can be performed efficiently on many samples. The advent of array CGH technology makes it possible to monitor DNA copy number changes on a genomic scale and many projects have been launched for studying the genome in specific diseases. [0177] In one embodiment, whole genome array-based comparative genome hybridization (array CGH) analysis, or array CGH on a subset of genomic regions, can be used to efficiently interrogate human genomes for genomic imbalances at multiple loci within a single assay. The development of comparative genomic hybridization (CGH) (Kallioniemi et al., Science. 258:818 (1992)) provided the first efficient approach to scanning entire genomes for variations in DNA copy number. The importance of normal copy number variation involving large segments of DNA has been unappreciated. Array CGH is a breakthrough technique in human genetics, which is attracting interest from clinicians working in fields as diverse as cancer and IVF (In Vitro Fertilization). The use of CGH microarrays in the clinic holds great promise for identifying regions of genomic imbalance associated with disease. Advances from identifying chromosomal critical regions associated with specific phenotypes to identifying the specific dosage sensitive genes can lead to therapeutic opportunities of benefit to patients. Array CGH is a specific, sensitive and rapid technique that can enable the screening of the whole genome in a single test. It can facilitate and accelerate the screening process in human genetics and is expected to have a profound impact on the screening and counseling of patients with genetic disorders. It is now possible to identify the exact location on the chromosome where an aberration has occurred and it is possible to map these changes directly onto the genomic sequence.

[0178] An array CGH approach provides a robust method for carrying out a genome-wide scan to find novel copy number variants (CNVs). The array CGH methods can use labeled fragments from a genome of interest, which can be competitively hybridized with a second differentially labeled genome to arrays that are spotted with cloned DNA fragments, revealing copy-number differences between the two genomes. Genomic clones (for example, BACs), cDNAs, PCR products and oligonucleotides, can all be used as array targets. The use of array CGH with BACs was one of the earliest employed methods and is popular, owing to the extensive coverage of the genome it provides, the availability of reliable mapping data and ready access to clones. The last of these factors is important both for the array experiments themselves, and for confirmatory FISH experiments.

[0179] In a typical CGH measurement, total genomic DNA is isolated from test and reference subjects; differentially labeled, and hybridized to a representation of the genome that allows the binding of sequences at different genomic locations to be distinguished. More than two genomes can be compared simultaneously with suitable labels.

Hybridization of highly repetitive sequences is typically suppressed by the inclusion of unlabeled Cot-1 DNA in the reaction. The relative hybridization intensity of the test and reference signals at a given location can be proportional to the relative copy number of those sequences in the test and reference genomes. If the reference genome is normal then increases and decreases in signal intensity ratios directly indicate DNA copy number variation within the test genome. Data are typically normalized so that the modal ratio for the genome is set to some standard value, typically 1.0 on a linear scale or 0.0 on a logarithmic scale. Additional measurements such as FISH or flow cytometry can be used to determine the actual-copy number associated with a ratio level.

[0180] In some embodiments, an array CGH procedure can include the following steps. First, large-insert clones, for example, BACs can be obtained from a supplier of clone libraries. Then, small amounts of clone DNA can be amplified, for example, by degenerate oligonucleotide-primed (DOP) PCR or ligation-mediated PCR in order to obtain sufficient quantities needed for spotting. Next, PCR products can be spotted onto glass slides using, for example, microarray robots equipped with high-precision printing pins. Depending on the number of clones to be spotted and the space available on the microarray slide, clones can either be spotted once per array or in replicate. Repeated spotting of the same clone on an array can increase precision of the measurements if the spot intensities are averaged, and allows for a detailed statistical analysis of the quality of the experiments. Subject and control DNAs can be labeled, for example, with either Cy3 or Cy5-dUTP using random priming and can be subsequently hybridized onto the microarray in a solution containing an excess of Cot1 -DNA to block repetitive sequences.

Hybridizations can either be performed manually under a coverslip, in a gasket with gentle rocking or, automatically using commercially available hybridization stations. These automated hybridization stations can allow for an active hybridization process, thereby improving the reproducibility as well as reducing the actual hybridization time, which increases throughput. The hybridized DNAs can detected through the two different fluorochromes using standard microarray scanning equipment with either a scanning confocal laser or a charge coupled device (CCD) camera-based reader, followed by spot identification using commercially or freely available software packages.

[0181] The use of CGH with arrays that comprise long oligonucleotides (60-100 bp) can improve the detection resolution (in some embodiments, as small as about 3-5 kb sized CNVs on arrays designed for interrogation of human whole genomes) over that achieved using BACs (limited to 50-100 kb or larger sized CNVs due to the large size of BAC clones). In some embodiments, the resolution of oligonucleotide CGH arrays is achieved via in situ synthesis of 1 -4 million unique features/probes per microarray, which can include microarrays available from Roche NimbleGen and Agilent Technologies. In addition to array CGH methods for copy number detecton, other embodiments for partial or whole genome analysis of CNVs within a genome include, but are not limited to, use of SNP genotyping microarrays and sequencing methods.

[0182] Another method for copy number detection that uses oligonucleotides can be representational oligonucleotide microarray analysis (ROMA). It is similar to that applied in the use of BAC and CGH arrays, but to increase the signal-to-noise ratio, the 'complexity' of the input DNA is reduced by a method called representation or whole-genome sampling. Here the DNA that is to be hybridized to the array can be treated by restriction digestion and then ligated to adapters, which results in the PCR-based amplification of fragments in a specific size-range. As a result, the amplified DNA can make up a fraction of the entire genomic sequence - that is, it is a representation of the input DNA that has significantly reduced complexity, which can lead to a reduction in background noise. Other suitable methods available to the skilled person can also be used, and are within scope of the present disclosure.

[0183] A comparison of one or more genomes relative to one or more other genomes with array CGH, or a variety of other CNV detection methods, can reveal the set of CNVs between two genomes, between one genome in comparison to multiple genomes, or between one set of genomes in comparison to another set of genomes. In some embodiments, an array CGH experiment can be performed by hybridizing a single test genome against a pooled sample of two or more genomes, which can result in minimizing the detection of higher frequency variants in the experiment. In some embodiments, a test genome can be hybridized alone (e.g., one-color detection) to a microarray, for example, using array CGH or SNP genotyping methods, and the comparison step to one or more reference genomes can be performed in silico to reveal the set of CNVs in the test genome relative to the one or more reference genomes. In one embodiment, a single test genome is compared to a single reference genome in a 2-color experiment wherein both genomes are cohybridized to the microarray.

[0184] Array CGH can be used to identify genes that are causative or associated with a particular phenotype, condition, or disease by comparing the set of CNVs found in the affected cohort to the set of CNVs found in an unaffected cohort. An unaffected cohort may consist of any individual unaffected by the phenotype, condition, or disease of interest, but in one embodiment is comprised of individuals or subjects that are apparently healthy (normal). Methods employed for such analyses are described in U.S. Pat. Nos.: 7,702,468 and 7,957,913. In some embodiments of CNV comparison methods, candidate genes that are causative or associated (i.e., potentially serving as a biomarker) with a phenotype, condition, or disease will be identified by CNVs that occur in the affected cohort but not in the unaffected cohort, or present at much lower frequency in the unaffected cohort as compared to the affected cohort. In another embodiment of CNV comparison methods, one or more CNVs may be present at much higher frequency in the unaffected cohort as compared to the affected cohort and thus may be indicative of protection for development of the disease or condition present in the affected cohort. In some embodiments of CNV comparison methods, candidate genes that are causative or associated (i.e., potentially serving as a biomarker) with a phenotype, condition, or disease will be identified by CNVs that occur at a statistically significant higher frequency in the affected cohort as compared their frequency in the unaffected cohort. Thus, CNVs detected in the affected cohort as compared to the unaffected cohort can serve as beacons of genes that are causative or associated with a particular phenotype, condition, or disease. In some embodiments, CNV detection and comparison methods can result in direct identification of the gene that is causative or associated with phenotype, condition, or disease if the CNVs are found to overlap with or encompass the gene(s). In some embodiments, CNV detection and comparison methods can result in identification of regulatory regions of the genome (e.g., promoters, enhancers, transcription factor binding sites) that regulate the expression of one or more genes that are causative or associated with the phenotype, condition, or disease of interest.

[0185] Due to the large amount of genetic variation between any two genomes, or two sets (cohorts) of genomes, being compared, one embodiment is to reduce the genetic variation search space by interrogating only CNVs, as opposed to the full set of genetic variants that can be identified in an individual's genome or exome. The set of CNVs that occur only, or at a statistically higher frequency, in the affected cohort as compared to the unaffected cohort can then be further investigated in targeted sequencing experiments to reveal the full set of genetic variants (of any size or type) that are causative or associated (e.g., potentially serving as a biomarker) with a phenotype, condition, or disease. It can be appreciated by those skilled in the art that the targeted sequencing experiments can be performed in both the affected and unaffected cohorts in order to identify the genetic variants (e.g., SNVs and indels) that occur only, or at a statistically significant higher frequency, in the affected individual or cohort as compared to the unaffected cohort. In another embodiment, the targeted sequencing experiments can be performed on the affected cohort and the variations found can be compared to public or private databases containing sequence variants present in unaffected subjects, or in some embodiments, the general population.

[0186] When investigating PD, it can be appreciated by those skilled in the art that the number of PD candidate genes (or regulatory sequences) identified via CNV (or other variant types) detection methods may increase or decrease when additional PD cohorts are analyzed. Similarly, the number of PD candidate genes (or regulatory sequences), for example, identified via CNV (or other variant types) detection methods may increase or decrease when additional unaffected cohorts are used to interpret the affected cohort CNVs (or other variant types). For very rare CNVs (e.g., <0.1% frequency in the general population), only a single case may be observed in a given PD cohort (e.g., 100 cases) but further statistical significance or evidence for the gene (or regulatory sequence/locus in the genome) can be established by: 1 ) CNV analysis of additional PD cohorts, 2) CNV analysis of additional Normal cohorts, 3) targeted gene sequencing of both PD and Normal cohorts, and/or 4) functional characterization of the PD candidate gene (e.g., in silico analysis of the predicted impact of the candidate mutation on the gene product, RNAi knockdown experiments, biochemical assays on PD patient tissue, gene expression analysis of disease-relevant tissues or of induced pluripotent stem cells (iPSCs) created from the PD patient(s) harboring the candidate PD-causing genetic variant).

[0187] It can be appreciated by those skilled in the art that a candidate gene may validate as causative of the phenotype, condition, or disease (e.g. , PD), which may, for example, be confirmed via mechanism of action experiments, or it may serve as a biomarker of the phenotype, condition, or disease. Thus, in the example of PD, in some embodiments, the PD -specific gene (or regulatory sequence/locus) may be a biomarker of age-of-onset for PD and disease severity, and thus have diagnostic utility for monitoring patients known to be at risk for PD or as a general screening test in the population for early diagnosis of the disease. In some embodiments, the PD-specific gene/biomarker may be an indicator of drug response (e.g., a particular subtype of PD may respond best to a therapeutic targeting a particular phenotype, causative gene, or other gene in the same pathway as the causative gene) and thus have utility during drug development in clinical trials. For example, clinical trials for a therapeutic that targets a PD genetic subtype comprising only 10% of all patients exhibiting symptoms of PD, can be designed to comprise only those 10% of patients with a specific genotype(s) in order to reduce the time and cost of such clinical trials (e.g., smaller number of patients in the clinical trial). It can be appreciated by those skilled in the art that such patient stratification methods (i.e., specific genotypes correlated with the disease or drug response) can be employed not only for targeted therapeutics, but in general for any drug that is approved or in development (i.e., the mechanism of action may or may not be known). For example, drugs in development or approved to treat, for example, cancer, may have utility in being repurposed to treat PD. Such patient stratification methods can also be utilized to develop a companion diagnostic test (e.g., comprising the specific genes/genotypes found in patients that are indicative of drug response) for a particular drug, either concurrently during the clinical trials for the drug or after drug approval (e.g., as a new indication or for the physician to use in guiding medical decisions for the patient).

[0188] Further links to PD pathology may be established via pathway analysis of the genes, which may take into consideration binding interactions (e.g., via yeast 2-hybrid screen) and molecular events (e.g., kinase activity or other enzymatic processes) if such information is available for the gene(s) of interest (e.g., specified in the analysis). Both commercial (e.g., Ingenuity's IPA software and Thomson Reuter's GeneGo software) and open source software (e.g., String: string-db.org/) are available for such analyses. To assess connections to established PD biology, analyses can be performed for the set of candidate PD genes independently or against known causative PD genes singly or as a group. For example, see Figure 10.

[0189] A method of screening a subject for a disease or disorder can comprise assaying a nucleic acid sample from the subject to detect sequence information for more than one genetic loci and comparing the sequence information to a panel of nucleic acid biomarkers and screening the subject for the presence or absence of PD if one or more of low frequency biomarkers in the panel are present in the sequence information. The panel may comprise at least one nucleic acid biomarker for each of the more than one genetic loci. For example, the panel can comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, 200 or more nucleic acid biomarkers for each of the more than one genetic loci. The panel may comprise at least 25 low frequency biomarkers. For example, the panel can comprise at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 135, 150, 175, 200, 250, 500, or 1000 or more low frequency biomarkers. A low frequency biomarker can occur at a frequency of 0.1 % or less in a population of subjects without a diagnosis of the disease or disorder. For example, a low frequency biomarker can occur at a frequency of 0.05%, 0.01 %, 0.005%, 0.001 %, 0.0005%, 0.0001 %, 0.00005%, or 0.00001 % or less in a population of subjects without a diagnosis of the disease or disorder.

[0190] In some embodiments, the presence or absence of PD in the subject can be determined with at least 50% confidence. For example, the presence or absence of the disease or disorder in the subject can be determined with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% confidence.

[0191] In one embodiment, PD candidate CNV subregions and genes associated with these regions may be determined or identified by comparing genetic data from a cohort of normal individuals (NVE) to that of a cohort of individuals known to have, or be susceptible to PD.

[0192] In some embodiments, genomic DNA samples from individuals within a

NVE cohort and/or a PD cohort can be considered test subject DNA samples and hybridized against one or more, sex-matched reference DNA samples from individuals. For example, reference DNA samples can be labeled with a fluorophore such as Cy5, using methods described herein, and test subject DNA samples can be labeled with a different fluorophore, such as Cy3. After labeling, samples can be combined and can be co- hybridized to a microarray and analyzed using any of the methods described herein, such as aCGH. Arrays can then be scanned and the data can be analyzed with software.

Genetic alterations, such as CNVs, can be called using any of the methods described herein. A list of the genetic alterations, such as CNVs, can be generated for each cohort. The list of CNVs can be used to generate a master list of non-redundant CNVs and/or CNV subregions for each cohort. The list can be based on the presence or absence of the CNV subregion in individuals within the cohort. In this manner, the master list can contain a number of distinct CNV subregions, some of which are uniquely present in a single individual and some of which are present in multiple individuals.

[0193] In some embodiments, CNV subregions of interest may be obtained by annotation of each CNV subregion with relevant information, such as overlap with known genes and/or exons. In some embodiments, CNV subregions of interest can be obtained by calculating the OR for a CNV subregion according to the following formula: OR = ( PD /((# individuals in PD cohort) - PD))/(NVE/((# individuals in NVE cohort) - NVE)), where: PD = number of PD individuals with a CNV subregion of interest and NVE = number of NVE individuals with the CNV subregion of interest. If NVE=0, it can be set to 1 to avoid dealing with infinities in cases where no CNVs are seen in the NVE.

[0194] The number of individuals in any given cohort may be at least about 10, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2500, 5000, 7500, 10,000, 100,000, or more.

[0195] In some embodiments, a CNV subregion/gene can be of interest if the CNV subregion overlaps a known gene, and is associated with an OR of at least 6, e.g., at least 35. For example, a CNV subregion/gene can be of interest if the CNV subregion overlaps a known gene, and is associated with an OR of at least 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, a CNV subregion/gene can be of interest if the CNV subregion overlaps a known gene, and is associated with an OR from about 6- 100, 6-50, 6-40, 6-30, 6-20, 6-10, 6-9, 6-8, 6-7, 8-100, 8-50, 8-40, 8-30, 8-20, 8-10, 10-100, 10-50, 10-40, 10-30, 10-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40- 50, 50-100, or 5-7. The CNV subregion/gene can be an exonic or intronic part of the gene, or both.

[0196] In some embodiments, a CNV subregion/gene can be of interest if the CNV subregion does not overlap a known gene (e.g., is non-genic or intergenic) and is associated with an OR of at least 4 or higher. For example, a CNV subregion/gene can be of interest if the CNV subregion does not overlap a known gene (e.g., is non-genic or intergenic) and is associated with an OR of at least 5, 6, 7, 9, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, a CNV subregion/gene can be of interest if the CNV subregion does not overlap a known gene (e.g., is non-genic or intergenic) and is associated with an OR from about 5-100, 5-50, 5-40, 5-30, 5-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50-100, or 9-11.

[0197] In some embodiments, a CNV subregion/gene can be of interest based on the OR associated with the sum of PD cases and the sum of NVE cases affecting the same gene (including distinct CNV subregions). For example, a CNV subregion/gene can be of interest if the OR associated with the sum of PD cases and the sum of NVE cases affecting the same gene (including distinct CNV subregions) is at least 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, a CNV subregion/gene can be of interest if the OR associated with the sum of PD cases and the sum of NVE cases affecting the same gene (including distinct CNV subregions) is from about 4-100, 4-50, 4- 40, 4-30, 4-20, 4-10, 4-9, 4-8, 4-7, 8-100, 8-50, 8-40, 8-30, 8-20, 8-10, 10-100, 10-50, 10- 40, 10-30, 10-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50- 100, or 5-7.

Computer-Implemented Aspects

[0198] As understood by those of ordinary skill in the art, the methods and information described herein (genetic variation association with PD) can be implemented, in all or in part, as computer executable instructions on known computer readable media. For example, the methods described herein can be implemented in hardware. Alternatively, the method can be implemented in software stored in, for example, one or more memories or other computer readable medium and implemented on one or more processors. As is known, the processors can be associated with one or more controllers, calculation units and/or other units of a computer system, or implanted in firmware as desired. If implemented in software, the routines can be stored in any computer readable memory such as in RAM, ROM, flash memory, a magnetic disk, a laser disk, or other storage medium, as is also known. Likewise, this software can be delivered to a computing device via any known delivery method including, for example, over a communication channel such as a telephone line, the Internet, a wireless connection, etc., or via a transportable medium, such as a computer readable disk, flash drive, etc.

[0199] More generally, and as understood by those of ordinary skill in the art, the various steps described above can be implemented as various blocks, operations, tools, modules and techniques which, in turn, can be implemented in hardware, firmware, software, or any combination of hardware, firmware, and/or software. When implemented in hardware, some or all of the blocks, operations, techniques, etc. can be implemented in, for example, a custom integrated circuit (IC), an application specific integrated circuit

(ASIC), a field programmable logic array (FPGA), a programmable logic array (PLA), etc.

[0200] Results from such genotyping can be stored in a data storage unit, such as a data carrier, including computer databases, data storage disks, or by other convenient data storage means. In certain embodiments, the computer database is an object database, a relational database or a post- relational database. Data can be retrieved from the data storage unit using any convenient data query method. [0201] When implemented in software, the software can be stored in any known computer readable medium such as on a magnetic disk, an optical disk, or other storage medium, in a RAM or ROM or flash memory of a computer, processor, hard disk drive, optical disk drive, tape drive, etc. Likewise, the software can be delivered to a user or a computing system via any known delivery method including, for example, on a computer readable disk or other transportable computer storage mechanism.

[0202] The steps of the claimed methods can be operational with numerous other general purpose or special purpose computing system environments or configurations. Examples of well-known computing systems, environments, and/or configurations that can be suitable for use with the methods or system of the claims include, but are not limited to, personal computers, server computers, hand-held or laptop devices, multiprocessor systems, microprocessor-based systems, set. top boxes, programmable consumer electronics, network PCs, minicomputers, mainframe computers, distributed computing environments that include any of the above systems or devices, and the like.

[0203] The steps of the claimed method and system can be described in the general context of computer-executable instructions, such as program modules, being executed by a computer. Generally, program modules include routines, programs, objects, components, and/or data structures that perform particular tasks or implement particular abstract data types. The methods and apparatus can also be practiced in distributed computing environments where tasks are performed by remote processing devices that are linked through a communications network. In both integrated and distributed computing environments, program modules can be located in both local and remote computer storage media including memory storage devices. Numerous alternative embodiments could be implemented, using either current technology or technology developed after the filing date of this application, which would still fall within the scope of the claims defining the disclosure.

[0204] While the risk evaluation system and method, and other elements, have been described as being implemented in software, they can be implemented in hardware, firmware, etc., and can be implemented by any other processor. Thus, the elements described herein can be implemented in a standard multi-purpose CPU or on specifically designed hardware or firmware such as an application-specific integrated circuit (ASIC) or other hard-wired device as desired. When implemented in. software, the software routine can be stored in any computer readable memory such as on a magnetic disk, a laser disk, or other storage medium, in a RAM or ROM of a computer or processor, in any database, etc. Likewise, this software can be delivered to a user or a screening system via any known or desired delivery method including, for example, on a computer readable disk or other transportable computer storage mechanism or over a communication channel, for example a telephone line, the internet, or wireless communication. Modifications and variations can be made in the techniques and structures described and illustrated herein without departing from the spirit and scope of the present disclosure.

PD Therapeutics

[0205] There is no cure for Parkinson's disease, but medications, surgery and multidisciplinary management can provide relief from the symptoms. Therapeutic options include L-dopa/DDC-inhibitors, COMT-inhibitors, e.g., entacapone or tolcapone, MAO-B- inhibitors, e.g., selegiline or rasagiline, NMDA-antagonists, e.g., amantadine or budipin, ergolin dopamine-agonists, e.g., bromocriptine, cabergoline, a-dihydroergocriptine, lisuride or pergolide, non-ergolin dopamine-agonists, e.g., apomorphine, piribedil, pramipexole, ropinirole or rotigotine, anti-depressives, e.g., SSRI (such as mirtazapine), anti-psychotics, e.g. , neuroleptics (clozapine or quetiapine), anti-dementia agents, e.g., AChEI (donepezil, rivastigmine, galantamine) or NMDA-antagonists (e.g., memantine), as well as pardoprunox (SLV 308), which is a partial D^agonist and full 5HT, A agonist, safinamide (PNU 151774E) which is a MAO-B/DA-reuptake/Glu-release inhibitor, AFQ056, which is a metabotropic- glutamate-receptor 5 antagonist, perampanel (E2007), which is a AMPA-glutamate-receptor antagonist, istradefylline (KW-6002), which is an adenosine A2a-receptor-antagonist or pitolisant (BF 2.649), which is histamine H3-antagonist. As disclosed herein, therapeutic options may be matched to each PD case based on one or more genetic variations, e.g., one or more CNVs, in each patient.

[0206] Thus, PD patients having a particular genetic variation may benefit from specific disease modifying therapies. For example for motor fluctuations, ACR 325, a dopamine-receptor stabilizer, AP09004(safety), a dual release gastric retentive, or XP21279, an L-dopa prodrug absorbed in the colon; for motor dysfunction, exenatide, a glucose metabolism/insulin regulator, nicotine, a nicotinic acetylcholine receptor agonist, PYM50028, an oral neurotrophic factor-inducing drug or V1512(levodopa methylester + carbidopa) may be useful or have improved efficacy in PD patients having a particular genetic variation. Others include caffeine, an adenosine receptor antagonist, for excessive daytime somnolence, fipamezole, an alpha-2 adrenergic antagonist, for orthostatic hypotension/LI D. Also for motor dysfunction, IPX066, an extended-release carbidopa-l- dopa, preladenant; for motor fluctuations, treating H. Pylori and apomorphinenasal powder/nasal spray ; for depression, citalopram, paroxetine or venlafaxine; for early cognitive impairment, donepezil (early dementia) or piribedil (vigilance and cognitive functions, may be useful or have improved efficacy in PD patients having a particular genetic variation. Others include eszopiclonefor insomnia, pimavanserintartrate (ACP-103) for psychosis, rivastigmine for apathy without dementia, desmopressin for nocturnal micturition frequency, lubiprostone for constipation, memantine for gait disorders and attention deficit, naltrexone for impulse control disorders, rasagiline for apathy, rasagiline for depression, rasagiline for hyposmia, rasagiline for sleep disturbances, and rivastigmine for dementia.

[0207] The main families of drugs useful for treating motor symptoms are dopamine oriented including levodopa (usually combined with a dopa decarboxylase inhibitor or COMT inhibitor), direct dopamine agonists, COMT inhibitor and MAO-B inhibitors. The stage of the disease determines which group is most useful. Two stages are usually distinguished: an initial stage in which the individual with PD has already developed some.disability for which he needs pharmacological treatment, then a second stage in which an individual develops motor complications related to levodopa usage. Treatment in the initial stage aims for an optimal tradeoff between good symptom control and side-effects resulting from enhancement of dopaminergic function. The start of levodopa (or L-DOPA) treatment may be delayed by using other medications such as MAO-B inhibitors and dopamine agonists, in the hope of delaying the onset of dyskinesias. In the second stage the aim is to reduce symptoms while controlling fluctuations of the response to medication. Sudden withdrawals from medication or overuse have to be managed. When medications are not enough to control symptoms, surgery and deep brain stimulation can be of use. In the final stages of the disease, palliative care is provided to enhance quality of life.

[0208] Levodopa has been the most widely used treatment for over 30 years. L- DOPA is converted into dopamine in the dopaminergic neurons by dopa decarboxylase. Since motor symptoms are produced by a lack of dopamine in the substantia nigra, the administration of L-DOPA temporarily diminishes the motor symptoms. Only 5-10% of L- DOPA crosses the blood-brain barrier. The remainder is often metabolized to dopamine elsewhere, causing a variety of side effects including nausea, dyskinesias and joint stiffness. Carbidopa and benserazide are peripheral dopa decarboxylase inhibitors, which help to prevent the metabolism of L-DOPA before it reaches the dopaminergic neurons, therefore reducing side effects and increasing bioavailability. They are generally given as combination preparations with levodopa. Existing preparations are carbidopa/levodopa (co- careldopa) and benserazide/levodopa (co-beneldopa). Levodopa has been related to dopamine dysregulation syndrome, which is a compulsive overuse of the medication, and punding. There are controlled release versions of levodopa in the form intravenous and intestinal infusions that spread out the effect of the medication. These slow-release levodopa preparations have not shown an increased control of motor symptoms or motor complications when compared to immediate release preparations.

[0209] Tolcapone inhibits the COMT enzyme, which degrades dopamine, thereby prolonging the effects of levodopa. It has been used to complement levodopa; however, its usefulness is limited by possible side effects such as liver damage. A similarly effective drug, entacapone, has not been shown to cause significant alterations of liver function. Licensed preparations of entacapone contain entacapone alone or in combination with carbidopa and levodopa.

[0210] Levodopa preparations lead in the long term to the development of motor complications characterized by involuntary movements called dyskinesias and fluctuations in the response to medication. When this occurs a person with PD can change from phases with good response to medication and few symptoms ("on" state), to phases with no response to medication and significant motor symptoms ("off" state). For this reason, levodopa doses are kept as low as possible while maintaining functionality. Delaying the initiation of therapy with levodopa by using alternatives (dopamine agonists and MAO-B inhibitors) is common practice. A former strategy to reduce motor complications was to withdraw L-DOPA medication for some time. This is discouraged now, since it can bring dangerous side effects such as neuroleptic malignant syndrome. Most people with PD eventually need levodopa and later develop motor side effects.

[0211] Several dopamine agonists that bind to dopaminergic post-synaptic receptors in the brain have similar effects to levodopa. These were initially used for individuals experiencing on-off fluctuations and dyskinesias as a complementary therapy to levodopa; they are now mainly used on their own as an initial therapy for motor symptoms with the aim of delaying motor complications. When used in late PD they are useful at reducing the off periods. Dopamine agonists include bromocriptine, pergolide, pramipexole, ropinirole, piribedil, cabergoline, apomorphine and lisuride.

[0212] Dopamine agonists produce significant, although usually mild, side effects including drowsiness, hallucinations, insomnia, nausea and constipation. Sometimes side effects appear even at a minimal clinically effective dose, leading the physician to search for a different drug. Compared with levodopa, dopamine agonists may delay motor complications of medication use but are less effective at controlling symptoms.

Nevertheless, they are usually effective enough to manage symptoms in the initial years. They tend to be more expensive than levodopa. Dyskinesias due to dopamine agonists are rare in younger people who have PD, but along with other side effects, become more common with age at onset. Thus dopamine agonists are the preferred initial treatment for earlier onset, as opposed to levodopa in later onset. Agonists have been related to impulse control disorders (such as compulsive sexual activity and eating, and pathological gambling and shopping) even more strongly than levodopa.

[0213] Apomorphine, a non-orally administered dopamine agonist, may be used to reduce off periods and dyskinesia in late PD. It is administered by intermittent injections or continuous subcutaneous infusions. Since secondary effects such as confusion and hallucinations are common, individuals receiving apomorphine treatment should be closely monitored. Two dopamine agonists that are administered through skin patches (lisuride and rotigotine) have been recently found to be useful for patients in initial stages and preliminary positive results has been published on the control of off states in patients in the advanced state.

[0214] MAO-B inhibitors (selegiline and rasagiline) increase the level of dopamine in the basal ganglia by blocking its metabolism. They inhibit monoamine oxidase-B (MAO- B) which breaks down dopamine secreted by the dopaminergic neurons. The reduction in MAO-B activity results in increased L-DOPA in the striatum. Like dopamine agonists, MAO- B inhibitors used as monotherapy improve motor symptoms and delay the need for levodopa in early disease, but produce more adverse effects and are less effective than levodopa. There are few studies of their effectiveness in the advanced stage, although results suggest that they are useful to reduce fluctuations between on and off periods. An initial study indicated that selegiline in combination with levodopa increased the risk of death, but this was later disproven.

[0215] Other drugs such as amantadine and anticholinergics may be useful as treatment of motor symptoms. However, the evidence supporting them lacks quality, so they are not first choice treatments. In addition to motor symptoms, PD is accompanied by a diverse range of symptoms. A number of drugs have been used to treat some of these problems. Examples are the use of clozapine for psychosis, cholinesterase inhibitors for dementia, and modaf inil for daytime sleepiness. A 2010 meta-analysis found that nonsteroidal anti-inflammatory drugs (apart from acetaminophen and aspirin), have been associated with at least a 15 percent (higher in long-term and regular users) reduction of incidence of the development of Parkinson's disease.

[0216] Treating motor symptoms with surgery was once a common practice, but since the discovery of levodopa, the number of operations declined. Studies in the past few decades have led to great improvements in surgical techniques, so that surgery is again being used in people with advanced PD for whom drug therapy is no longer sufficient. Surgery for PD can be divided in two main groups: lesional and deep brain stimulation (DBS). Target areas for DBS or lesions include the thalamus, the globus pallidus or the subthalamic nucleus. Deep brain stimulation (DBS) is the most commonly used surgical treatment. It involves the implantation of a medical device called a brain pacemaker, which sends electrical impulses to specific parts of the brain. DBS is recommended for people who have PD who suffer from motor fluctuations and tremor inadequately controlled by medication, or to those who are intolerant to medication, as long as they do not have severe neuropsychiatric problems. Other, less common, surgical therapies involve the formation of lesions in specific subcortical areas (a technique known as pallidotomy in the case of the lesion being produced in the globus pallidus). [0217] There is some evidence that speech or mobility problems can improve with rehabilitation, although studies are scarce and of low quality. Regular physical exercise with or without physiotherapy can be beneficial to maintain and improve mobility, flexibility, strength, gait speed, and quality of life. However, when an exercise program is performed under the supervision of a physiotherapist, there are more improvements in motor symptoms, mental and emotional functions, daily living activities, and quality of life compared to a self-supervised exercise program at home. In terms of improving flexibility and range of motion for patients experiencing rigidity, generalized relaxation techniques such as gentle rocking have been found to decrease excessive muscle tension. Other effective techniques to promote relaxation include slow rotational movements of the extremities and trunk, rhythmic initiation, diaphragmatic breathing, and meditation techniques. As for gait and addressing the challenges associated with the disease such as hypokinesia (slowness of movement), shuffling and decreased arm swing; physiotherapists have a variety of strategies to improve functional mobility and safety. Areas of interest with respect to gait during rehabilitation programs focus on but are not limited to improving gait speed, base of support, stride length, trunk and arm swing movement. Strategies include utilizing assistive equipment (pole walking and treadmill walking), verbal cueing (manual, visual and auditory), exercises (marching and PNF patterns) and altering environments (surfaces, inputs, open vs. closed). Strengthening exercises have shown improvements in strength and motor function for patients with primary muscular weakness and weakness related to inactivity with mild to moderate Parkinson's disease. However, reports show a significant interaction between strength and the time the medications was taken. Therefore, it is recommended that patients should perform exercises 45 minutes to one hour after medications, when the patient is at their best. Also, due to the forward flexed posture, and respiratory dysfunctions in advanced Parkinson's disease, deep diaphragmatic breathing exercises are beneficial in improving chest wall mobility and vital capacity. Exercise may improve constipation.

[0218] One of the most widely practiced treatments for speech disorders associated with Parkinson's disease is the Lee Silverman voice treatment (LSVT). Speech therapy and specifically LSVT may improve speech. Occupational therapy (OT) aims to promote health and quality of life by helping people with the disease to participate in as many of their daily living activities as possible. There have been few studies on the effectiveness of OT and their quality is poor, although there is some indication that it may improve motor skills and quality of life for the duration of the therapy.

[0219] Muscles and nerves that control the digestive process may be affected by

PD, resulting in constipation and gastroparesis (food remaining in the stomach for a longer period of time than normal). A balanced diet, based on periodical nutritional assessments, is recommended and should be designed to avoid weight loss or gain and minimize consequences of gastrointestinal dysfunction. As the disease advances, swallowing difficulties (dysphagia) may appear. In such cases it may be helpful to use thickening agents for liquid intake and an upright posture when eating, both measures reducing the risk of choking. Gastrostomy to deliver food directly into the stomach is possible in severe cases.

[0220] Levodopa and proteins use the same transportation system in the intestine and the blood-brain barrier, thereby competing for access. When they are taken together, this results in a reduced effectiveness of the drug. Therefore, when levodopa is introduced, excessive protein consumption is discouraged and well balanced Mediterranean diet is recommended. In advanced stages, additional intake of low-protein products such as bread or pasta is recommended for similar reasons. To minimize interaction with proteins, levodopa should be taken 30 minutes before meals. At the same time, regimens for PD restrict proteins during breakfast and lunch, allowing protein intake in the evening. A person skilled in the art will appreciate and understand that the genetic variants described herein in general may not, by themselves, provide an absolute identification of individuals who can develop a ND or related conditions. The variants described herein can indicate increased and/or decreased likelihood that individuals carrying the at-risk or protective variants of the disclosure can develop symptoms associated with a ND. This information can be used to, for example, initiate preventive measures at an early stage, perform regular physical and/or mental exams to monitor the progress and/or appearance of symptoms, or to schedule exams at a regular interval to identify early symptoms, so as to be able to apply treatment at an early stage. This is in particular important since NDs and related disorders are heterogeneous disorders with symptoms that can be individually vague. Screening criteria can comprise a number of symptoms to be present over a period of time; therefore, it is important to be able to establish additional risk factors that can aid in the screening, or facilitate the screening through in-depth phenotyping and/or more frequent examination, or both. For example, individuals with early symptoms that typically are not individually associated with a clinical screening of a ND and carry an at-risk genetic variation can benefit from early therapeutic treatment, or other preventive measure, or more rigorous supervision or more frequent examination. Likewise, individuals that have a family history of the disease, or are carriers of other risk factors associated with a ND can, in the context of additionally carrying at least one at-risk genetic variation, benefit from early therapy or other treatment.

[0221] Early symptoms of behavioral disorders such as a ND and related conditions may not be sufficient to fulfill standardized screening criteria. To fulfill those, a certain pattern of symptoms and behavioral disturbance needs to manifest itself over a period of time. Sometimes, certain physical characteristics can also be present. This makes at-risk genetic variants valuable in a screening setting, in particular high-risk variants. Determination of the presence of such variants warrants increased monitoring of the individual in question. Appearance of symptoms combined with the presence of such variants facilitates early screening, which makes early treatment possible. Genetic testing can thus be used to aid in the screening of disease in its early stages, before all criteria for formal screening criteria are all fulfilled. It is well established that early treatment is extremely important for NDs and related disorders, which lends further support to the value of genetic testing for early diagnosis, prognosis, or theranosis of these disorders.

[0222] Disease modifying (neuroprotective therapies) that may be useful or have improved efficacy in PD patients having a particular genetic variation include those for oxidative stress, e.g., antioxidants (e.g., Vitamin E, Vitamin C, Fe chelators), for mitochondrial dysfunction, e.g., bioenergetics substances (e.g., CoQ10), for excitotoxicity, e.g., NMDA antagonists (e.g., MK801 ), for inflammation, e.g., anti-inflammatory substances (e.g., Cox 2 inhibitors), for protein-degradation, e.g., proteasomal enhancer, such as rapamycin, fpr neuronal dysfunction, e.g., trophic factors (for instance, GDNF, nurturing, neuregulin), and for apoptosis, anti-apoptotic substances (e.g., DA agonists, AKT, caspase inhibitors). See Table 4.

Table 4

[0223] Exemplary neuroprotective therapies include but are not limited to

Coenzyme Q10, a mitochondrial enhancer, GPI 1485, a novel immunophilin compound, deferiprone, an iron chelator, green tea polyphenols, antioxidants, inosine, a nucleoside, isradipine, a calcium channel blocker, mitoquinone, a mitochondrial antioxidant, exenatide, a glucose metabolism / insulin regulator, paliroden(SR57667B), minocycline, a tetracycline, creatine, a dietary supplement, nicotine, a nicotinic acetylcholine receptor agonist, granulocyte-CSF, a hematopoietic growth factor, PY 50028, an oral neurotrophic factor inducer, sNN0031 , intracerebroventricular platelet derived growth factor, SPM962, creatine, a dietary supplement, rasagiline, a monoamine oxidase-B inhibitor, ProSavin, a lentivector delivery system to transfer three genes, aromatic amino acid dopa decarboxylase, tyrosine hydroxylase and GTP-cyclohydrolase 1 , to the striatum, reprogramming transduced cells to secrete dopamine, GAD-gene therapy, adeno-associated virus delivery of aglutamic acid decarboxylase gene to the subthalamic nucleus, CERE-120, an adeno-associated virus serotype 2 (AAV2) delivery of the gene for the neurotrophin neurturin, or stem cell therapy.

[0224] The present disclosure provides methods for matching compounds or agents and a subject having a ND and one or more specific genetic variations. The genetic variations and associated proteins of the disclosure are also useful as targets for the identification and/or development of therapeutic agents. In certain embodiments, such methods include assaying the ability of an agent or compound to modulate the activity and/or expression of a nucleic acid that is associated with at least one genetic variation described herein, encoded products of the gene sequence, and any other molecules or proteins associated with these genes. This in turn can be used to identify agents or compounds that inhibit, enhance, or alter the undesired activity, localization, binding and/or expression of the encoded nucleic acid product, such as mRNA or polypeptides. The genes associated with the CNVs are shown in Figures 10A-D. Assays for performing such experiments can be performed in cell-based systems or in cell-free systems, as known to the skilled person. Cell-based systems include cells naturally expressing the nucleic acids of interest, or recombinant cells that have been genetically modified so as to express a certain desired nucleic acid molecule.

[0225] Variant gene expression in a subject can be assessed by expression of a variant-containing nucleic acid sequence or by altered expression of a normal/wild-type nucleic acid sequence due to variants affecting the level or pattern of expression of the normal transcripts, for example variants in the regulatory or control region of the gene. Assays for gene expression include direct nucleic acid assays (m RNA), assays for expressed protein levels, or assays of collateral compounds involved in a pathway, for example a signal pathway. Furthermore, the expression of genes that are up- or down- regulated in response to the signal pathway can also be assayed. One embodiment includes operably linking a reporter gene, such as luciferase, to the regulatory region of one or more gene of interest.

[0226] Modulators of gene expression can in some embodiments be identified when a cell is contacted with a candidate compound or agent, and the expression of mRNA is determined. The expression level of mRNA in the presence of the candidate compound or agent is compared to the expression level in the absence of the compound or agent. Based on this comparison, candidate compounds or agents for treating a ND can be identified as those modulating the gene expression of the variant gene, or gene expression of one or more other genes occurring within the same biological pathway or known, for example, to be binding partners of the variant gene. When expression of mRNA or the encoded protein is statistically significantly greater in the presence of the candidate compound or agent than in its absence, then the candidate compound or agent is identified as a stimulator or up-regulator of expression of the nucleic acid. When nucleic acid expression or protein level is statistically significantly less in the presence of the candidate compound or agent than in its absence, then the candidate compound can be identified as an inhibitor or down-regulator of the nucleic acid expression. The disclosure further provides methods of treatment using a compound identified through drug (compound and/or agent) screening as a gene modulator.

[0227] The genetic variations described herein can be used to identify novel therapeutic targets for a ND. For example, genes containing, or in linkage disequilibrium with, the genetic variations, or their products, as well as genes or their products that are directly or indirectly regulated by or interact with these variant genes or their products, can be targeted for the development of therapeutic agents to treat a ND, or prevent or delay onset of symptoms associated with a ND. Therapeutic agents can comprise one or more of, for example, small non-protein and non- nucleic acids, proteins, peptides, protein fragments, nucleic acids (DNA, RNAJ, PNA (peptide nucleic acids)), or their derivatives or mimetics which can modulate the function and/or levels of the target genes or their gene products. In some embodiments, treatment of PD can comprise treatment of one of the genes, or gene products derived thereof, such as mRNA or a polypeptide, with one or more of the therapeutics disclosed herein. In some embodiments, treatment of PD can comprise treatment of 2 or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10 or more of the genes, or gene products derived there from, with 2 or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10 or more of the therapeutics disclosed herein.

RNA therapeutics

[0228] The nucleic acids and/or variants of the disclosure, or nucleic acids comprising their complementary sequence, can be used as antisense constructs to control gene expression in cells, tissues or organs. The methodology associated with antisense techniques is well known to the skilled artisan, and is described and reviewed in Antisense Drug Technology: Principles, Strategies, and Applications, Crooke, Marcel Dekker Inc., New York (2001 ) In general, antisense nucleic acids are designed to be complementary to a region of mRNA expressed by a gene, so that the antisense molecule hybridizes to the mRNA, thus blocking translation of the mRNA into protein Several classes of antisense oligonucleotide are known to those skilled in the art, including cleavers and blockers. The former bind to target RNA sites, activate intracellular nucleases (e.g., Rnase H or Rnase L) that cleave the target RNA. Blockers bind to target RNA, inhibit protein translation by steric hindrance of the ribosomes. Examples of blockers include nucleic acids, morpholino compounds, locked nucleic acids and methylphosphonates (Thompson, Drug Discovery Today, 7:912 (2002)) Antisense oligonucleotides are useful directly as therapeutic agents, and are also useful for determining and validating gene function, for example by gene knock-out or gene knock-down experiments. Antisense technology is further described in Lavery et al., Curr. Qpin. Drug Discov. Devel.. 6:561 (2003), Stephens et al., Curr. Opin. Mol Ther., 5:118 (2003), Kurreck, Eur. J. Biochem.. 270:1628 (2003), Dias et al, Mol. Cancer Ter..1:347 (2002), Chen, Methods Mol. Med.. 7.5:621 (2003), Wang et al., Curr. Cancer Drug Targets.1:177 (2001 ), and Bennett, Antisense Nucleic Acid Drug. Dev.. 12:215 (2002).

[0229] The variants described herein can be used for the selection and design of antisense reagents that are specific for particular variants (e.g., particular genetic variations or polymorphic markers in linkage disequilibrium with particular genetic variations). Using information about the variants described herein, antisense oligonucleotides or other antisense molecules that specifically target mRNA molecules that contain one or more variants of the disclosure can be designed. In this manner, expression of mRNA molecules that contain one or more variants of the present disclosure (markers and/or haplotypes) can be inhibited or blocked. In some embodiments, the antisense molecules are designed to specifically bind a particular allelic form (i.e., one or several variants (alleles and/or haplotypes)) of the target nucleic acid, thereby inhibiting translation of a product originating from this specific allele or haplotype, but which do not bind other or alternate variants at the specific polymorphic sites of the target nucleic acid molecule.

[0230] As antisense molecules can be used to inactivate mRNA so as to inhibit gene expression, and thus protein expression, the molecules can be used to treat PD. The methodology can involve cleavage by means of ribozymes containing nucleotide sequences complementary to one or more regions in the mRNA that attenuate the ability of the mRNA to be translated. Such mRNA regions include, for example, protein-coding regions, in particular protein-coding regions corresponding to catalytic activity, substrate and/or ligand binding sites, or other functional domains of a protein.

[0231] The phenomenon of RNA interference (RNAi) has been actively studied for the last decade, since its original discovery in C. elegans (Fire et al., Nature. 391:806 (1998)), and in recent years its potential use in treatment of human disease has been actively pursued (reviewed in Kim & Rossi, Nature Rev. Genet.. 8:173 (2007)). RNA interference (RNAi), also called gene silencing, is based on using double-stranded RNA molecules (dsRNA) to turn off specific genes. In the cell, cytoplasmic double-stranded RNA molecules (dsRNA) are processed by cellular complexes into small interfering RNA (siRNA). The siRNA guide the targeting of a protein-RNA complex to specific sites on a target mRNA, leading to cleavage of the mRNA (Thompson, Drug Discovery Today. 7:912 (2002)). The siRNA molecules are typically about 20, 21 , 22 or 23 nucleotides in length. Thus, one aspect of the disclosure relates to isolated nucleic acid sequences, and the use of those molecules for RNA interference, for example as small interfering RNA molecules (siRNA). In some embodiments, the isolated nucleic acid sequences can be 18-26 nucleotides in length, or 19-25 nucleotides in length, or 20-24 nucleotides in length, or 21 , 22 or 23 nucleotides in length.

[0232] Another pathway for RNAi-mediated gene silencing originates in endogenously encoded primary microRNA (pn-miRNA) transcripts, which are processed in the cell to generate precursor miRNA (pre-miRNA). These miRNA molecules are exported from the nucleus to the cytoplasm, where they undergo processing to generate mature miRNA molecules (miRNA), which direct translational inhibition by recognizing target sites in the 3' untranslated regions of mRNAs, and subsequent mRNA degradation by processing P-bodies (reviewed in Kim & Rossi, Nature Rev. Genet.. 8:173 (2007)).

[0233] Clinical applications of RNAi include the incorporation of synthetic siRNA duplexes, which are approximately 20-23 nucleotides in size, and may have 3' overlaps of 2 nucleotides. Knockdown of gene expression is established by sequence-specific design for the target mRNA. Several commercial sites for optimal design and synthesis of such molecules are known to those skilled in the art. [0234] Other applications provide longer siRNA molecules (typically 25-30 nucleotides in length, such as about 27 nucleotides), as well as small hairpin RNAs (shRNAs; typically about 29 nucleotides in length). The latter are naturally expressed, as described in Amarzguioui et al. (FEBS Lett.. 579:5974 (2005)). Chemically synthetic siRNAs and shRNAs are substrates for in vivo processing, and in some cases provide more potent gene-silencing than shorter designs (Kim et al., Nature Biotechnol.. 23:222 (2005); Siola et al., Nature Biotechnol., 23:227 (2005)). In general siRNAs provide for transient silencing of gene expression, because their intracellular concentration is diluted by subsequent cell divisions. By contrast, expressed shRNAs mediate long-term, stable knockdown of target transcripts, for as long as transcription of the shRNA takes place

(Marques et al., Nature Biotechnol., 23:559 (2006), Brummelkamp et al., Science. 296:550 (2002)).

[0235] Since RNAi molecules, including siRNA, miRNA and shRNA, act in a sequence-dependent manner, variants described herein can be used to design RNAi reagents that recognize specific nucleic acids comprising specific genetic variations, alleles and/or haplotypes, while not recognizing nucleic acid sequences not comprising the genetic variation, or comprising other alleles or haplotypes. These RNAi reagents can thus recognize and destroy the target nucleic acid sequences. As with antisense reagents, RNAi reagents can be useful as therapeutic agents (i.e., for turning off disease- associated genes or disease-associated gene variants), but can also be useful for characterizing and validating gene function (e.g., by gene knock-out or gene knock-down experiments).

[0236] Delivery of RNAi can be performed by a range of methodologies known to those skilled in the art. Methods utilizing non-viral delivery include cholesterol, stable nucleic acid-lipid particle (SNALP), heavy-chain antibody fragment (Fab), aptamers and nanoparticles Viral delivery methods include use of lentivirus, adenovirus and adeno- associated virus The siRNA molecules are in some embodiments chemically modified to increase their stability. This can include modifications at the 2' position of the ribose, including 2'-0-methylpunnes and 2'- fluoropyrimidmes, which provide resistance to RNase activity. Other chemical modifications are possible and known to those skilled in the art.

[0237] The following references provide a further summary of RNAi, and possibilities for targeting specific genes using RNAi : Kim & Rossi. Nat. Rev. Genet.. 8:173 (2007), Chen & Rajewsky, Nat. Rev. Genet.. 8:93 (2007), Reynolds, et al. , Nat. Biotechnol., 22:326 (2004), Chi et al., Proc. Natl. Acad. Sa. USA. 100:6343 (2003), Vickers et al., J. Biol. Chem., 278:7108 (2003), Agami, Curr. Opin. Chem. Biol.. 6:829 (2002), Lavery, et al., Curr. Opin. Drug Discov. Devel.. 6:561 (2003), Shi, Trends Genet.. 19:9 (2003), Shuey et al.,

Drug Discov. Today. 7:1040 (2002), McManus et al., Nat. Rev. Genet.. 3:737 (2002), Xia et al., Nat. Biotechnol.. 20:1006 (2002), Plasterk et al., Curr. Qpin Genet. Dev.. 10:562 (2000), Bos er et al., Nat. Cell Biol., 2:E31 (2000), and Hunter, Curr. Biol., 9:R440 (1999).

[0238] A genetic defect leading to increased predisposition or risk for development of PD or a defect causing the disease, can be corrected permanently by administering to a subject carrying the defect a nucleic acid fragment that incorporates a repair sequence that supplies the normal/wild-type nucleotide(s) at the site of the genetic defect. Such site- specific repair sequence can encompass an RNA/DNA oligonucleotide that operates to promote endogenous repair of a subject's genomic DNA. The administration of the repair ' sequence can be performed by an appropriate vehicle, such as a complex with polyethelamine, encapsulated in anionic liposomes, a viral vector such as an adenovirus vector, or other pharmaceutical compositions suitable for promoting intracellular uptake of the administered nucleic acid The genetic defect can then be overcome, since the chimeric oligonucleotides induce the incorporation of the normal sequence into the genome of the subject, leading to expression of the normal/wild-type gene product. The replacement is propagated, thus rendering a permanent repair and alleviation of the symptoms associated with the disease or condition.

[0239] Double stranded oligonucleotides are formed by the assembly of two distinct oligonucleotide sequences where the oligonucleotide sequence of one strand is complementary to the oligonucleotide sequence of the second strand; such double stranded oligonucleotides are generally assembled from two separate oligonucleotides (e.g., siRNA), or from a single molecule that folds on itself to form a double stranded structure (e.g., shRNA or short hairpin RNA). These double stranded oligonucleotides known in the art all have a common feature in that each strand of the duplex has a distinct nucleotide sequence, wherein only one nucleotide sequence region (guide sequence or the antisense sequence) has complementarity to a target nucleic acid sequence and the other strand

(sense sequence) comprises nucleotide sequence that is homologous to the target nucleic acid sequence.

[0240] Double stranded RNA induced gene silencing can occur on at least three different levels: (i) transcription inactivation, which refers to RNA guided DNA or histone methylation; (ii) siRNA induced m RNA degradation; and (iii) mRNA induced transcriptional attenuation. It is generally considered that the major mechanism of RNA induced silencing (RNA interference, or RNAi) in mammalian cells is mRNA degradation. RNA interference (RNAi) is a mechanism that inhibits gene expression at the stage of translation or by hindering the transcription of specific genes. Specific RNAi pathway proteins are guided by the dsRNA to the targeted messenger RNA (mRNA), where they "cleave" the target, breaking it down into smaller portions that can no longer be translated into protein. Initial attempts to use RNAi in mammalian cells focused on the use of long strands of dsRNA. However, these attempts to induce RNAi met with limited success, due in part to the induction of the interferon response, which results in a general, as opposed to a target- specific, inhibition of protein synthesis. Thus, long dsRNA is not a viable option for RNAi in mammalian systems. Another outcome is epigenetic changes to a gene - histone modification and DNA methylation - affecting the degree the gene is transcribed.

[0241] More recently it has been shown that when short (18-30 bp) RNA duplexes are introduced into mammalian cells in culture, sequence-specific inhibition of target mRNA can be realized without inducing an interferon response. Certain of these short dsRNAs, referred to as small inhibitory RNAs ("siRNAs"), can act catalytically at sub-molar concentrations to cleave greater than 95% of the target mRNA in the cell. A description of the mechanisms for siRNA activity, as well as some of its applications are described in Provost et al., EMBO J„ £1:5864 (2002); Tabara et al., Cell, 109:861 (2002); Martinez et al. , Cell. 110:563 (2002); Hutvagner & Zamore, Science. 297:2056 (2002).

[0242] From a mechanistic perspective, introduction of long double stranded RNA into plants and invertebrate cells is broken down into siRNA by a Type III endonuclease known as Dicer. Sharp, RNA interference-2001 , Genes Dev.. 15:485 (2001 ). Dicer, a ribonuclease-lll-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3' overhangs. Bernstein, Caudy, Hammond, & Hannon, Nature, 409:363 (2001 ). The siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, Haley, & Zamore, Cell, 107:309 (2001 )). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleaves the target to induce silencing. Elbashir, Lendeckel, & Tuschl, Genes Dev., 15:188 (2001 ).

[0243] Generally, the antisense sequence is retained in the active RISC complex and guides the RISC to the target nucleotide sequence by means of complementary base- pairing of the antisense sequence with the target sequence for mediating sequence-specific RNA interference. It is known in the art that in some cell culture systems, certain types of unmodified siRNAs can exhibit "off target" effects. It is hypothesized that this off-target effect involves the participation of the sense sequence instead of the antisense sequence of the siRNA in the RISC complex (see for example, Schwarz et al., Cell, 115:199 (2003)). In this instance the sense sequence is believed to direct the RISC complex to a sequence (off- target sequence) that is distinct from the intended target sequence, resulting in the inhibition of the off-target sequence. In these double stranded nucleic acid sequences, each strand is complementary to a distinct target nucleic acid sequence. However, the off-targets that are affected by these dsRNAs are not entirely predictable and are non-specific. [0244] The term "siRNA" refers to small inhibitory RNA duplexes that induce the

RNA interference (RNAi) pathway. These molecules can vary in length (generally between 18-30 basepairs) and contain varying degrees of complementarity to their target mRNA in the antisense strand. Some, but not all, siRNA have unpaired overhanging bases on the 5' or 3' end of the sense strand and/or the antisense strand. The term "siRNA" includes duplexes of two separate strands, as well as single strands that can form hairpin structures comprising a duplex region. Small interfering RNA (siRNA), sometimes known as short interfering RNA or silencing RNA, are a class of 20-25 nucleotide-long double-stranded RNA molecules that play a variety of roles in biology.

[0245] While the two RNA strands do not need to be completely complementary, the strands should be sufficiently complementary to hybridize to form a duplex structure. In some instances, the complementary RNA strand can be less than 30 nucleotides, less than 25 nucleotides in length, about 19 to 24 nucleotides in length, or 20-23 nucleotides in length, including 22 nucleotides in length. The dsRNA of the present disclosure can further comprise at least one single-stranded nucleotide overhang. The dsRNA of the present disclosure can further comprise a substituted or chemically modified nucleotide. As discussed in detail below, the dsRNA can be synthesized by standard methods known in the art.

[0246] siRNA can be divided into five (5) groups including non-functional, semi- functional, functional, highly functional, and hyper-functional based on the level or degree of silencing that they induce in cultured cell lines. As used herein, these definitions are based on a set of conditions where the siRNA is transfected into the cell line at a concentration of 100 n and the level of silencing is tested at a time of roughly 24 hours after transfection, and not exceeding 72 hours after transfection. In this context, "non-functional siRNA" are defined as those siRNA that induce less than 50% (<50%) target silencing. "Semifunctional siRNA" induce 50-79% target silencing. "Functional siRNA" are molecules that induce 80-95% gene silencing. "Highly-functional siRNA" are molecules that induce greater than 95% gene silencing. "Hyperfunctional siRNA" are a special class of molecules. For purposes of this document, hyperfunctional siRNA are defined as those molecules that: (1 ) induce greater than 95% silencing of a specific target when they are transfected at subnanomolar concentrations (i.e., less than one nanomolar); and/or (2) induce functional (or better) levels of silencing for greater than 96 hours. These relative functionalities (though not intended to be absolutes) can be used to compare siRNAs to a particular target for applications such as functional genomics, target identification and therapeutics.

[0247] microRNAs (miRNA) are single-stranded RNA molecules of about 21 -23 nucleotides in length, which regulate gene expression. miRNAs are encoded by genes that are transcribed from DNA but not translated into protein (non-coding RNA) ; instead they are processed from primary transcripts known as pri-miRNA to short stem-loop structures called pre-miRNA and finally to functional miRNA. Mature miRNA molecules are partially complementary to one or more messenger RNA (mRNA) molecules, and their main function is to downregulate gene expression.

Antibody-Based Therapeutics

[0248] The present disclosure embodies agents that modulate a peptide sequence or RNA expressed from a gene associated with PD. The term biomarker, as used herein, can comprise a genetic variation of the present disclosure or a gene product, for example, RNA and polypeptides, of any one of the genes listed in Figures 8-11. Such modulating agents include, but are not limited to, proteins, peptides, peptidomimetics, peptoids, or any other forms of a molecule, which bind to, and alter the signaling or function associated with the PD associated biomarker, have an inhibitory or stimulatory effect on the PD associated biomarkers, or have a stimulatory or inhibitory effect on the expression or activity of the PD associated biomarkers' ligands, for example, polyclonal antibodies and/or monoclonal antibodies that specifically bind one form of the gene product but not to the other form of the gene product are also provided, or which bind a portion of either the variant or the reference gene product that contains the polymorphic site or sites.

[0249] In some embodiments, the present disclosure provides antibody-based agents targeting PD associated biomarkers. The antibody-based agents in any suitable form of an antibody, e.g., monoclonal, polyclonal, or synthetic, can be utilized in the therapeutic methods disclosed herein. The antibody-based agents include any target- binding fragment of an antibody and also peptibodies, which are engineered therapeutic molecules that can bind to human drug targets and contain peptides linked to the constant domains of antibodies. In some embodiments, the antibodies used for targeting PD associated biomarkers are humanized antibodies. Methods for humanizing antibodies are well known in the art. In some embodiments, the therapeutic antibodies comprise an antibody generated against PD associated biomarkers described in the present disclosure, wherein the antibodies are conjugated to another agent or agents, for example, a cytotoxic agent or agents.

[0250] The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain antigen-binding sites that specifically " bind an antigen. A molecule that specifically binds to a polypeptide of the disclosure is a molecule that binds to that polypeptide or a fragment thereof, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin. The disclosure provides polyclonal and monoclonal antibodies that bind to a polypeptide of the disclosure. The term "monoclonal antibody" or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of a polypeptide of the disclosure. A monoclonal antibody composition thus typically displays a single binding affinity for a particular polypeptide of the disclosure with which it immunoreacts.

[0251] Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a desired immunogen, e.g., polypeptide of the disclosure or a fragment thereof. The antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide. If desired, the antibody molecules directed against the polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, e.g., when the antibody titers are highest, antibody- producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein, Nature, 256:495 (1975), the human B cell hybridoma technique (Kozbor et al., Immunol. Today, 4:72 (1983)), the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy. Alan R. Liss (1985) Inc., pp. 77-96) or trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology ( 994) Coligan et al., (eds.) John Wiley & Sons, Inc., New York, NY). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds a polypeptide of the disclosure.

[0252] Any of the many well-known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating a monoclonal antibody to a polypeptide of the disclosure (see, e.g. , Current Protocols in Immunology, supra; Galfre et al., Nature, 266:55052 (1977) ; R.H. Kenneth, in Monoclonal Antibodies: A New

Dimension In Biological Analyses. Plenum Publishing Corp., New York, New York (1980) ; and Lerner, J. Biol. Med., 54:387 (1981 )). Moreover, the ordinarily skilled worker can appreciate that there are many variations of such methods that also would be useful.

Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody to a polypeptide of the disclosure can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide to thereby isolate immunoglobulin library members that bind the polypeptide. Kits for generating and screening phage display libraries are commercially available (e.g. , the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01 ; and the Stratagene SurfZAP a Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication Nos. WO 92/18619, WO 91/17271 , WO 92/20791 , WO 92/15679; WO 93/01288, WO

92/01047, WO 92/09690, and WO 90/02809; Fuchs et al., Biotechnology. 9:1370 (1991 ); Hay et al., Hum. Antibod. Hvbndomas. 3:81 (1992); Huse et al., Science. 246:1275 (1989); and Griffiths et al., EMBO J.. 12:725 (1993).

[0253] Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the disclosure. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art.

[0254] In general, antibodies of the disclosure (e.g., a monoclonal antibody) can be used to isolate a polypeptide of the disclosure by standard techniques, such as affinity chromatography or immunoprecipitation. A polypeptide-specific antibody can facilitate the purification of natural polypeptide from cells and of recombinants produced polypeptide expressed in host cells Moreover, an antibody specific for a polypeptide of the disclosure can be used to detect the polypeptide (e.g., in a cellular lysate, cell supernatant, or tissue sample) in order to evaluate the abundance and pattern of expression of the polypeptide. Antibodies can be used diagnostically, prognostically, or theranostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. The antibody can be coupled to a detectable substance to facilitate its detection. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein

isothiocyanate, rhodamine, dichlorotnazinylamine fluorescein, dansyl chloride or phycoerythnn; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 l, 31 1, 35 S or 3 H. Antibodies can also be useful in pharmacogenomic analysis. In such embodiments, antibodies against variant proteins encoded by nucleic acids according to the disclosure, such as variant proteins that are encoded by nucleic acids that contain at least one genetic variation of the disclosure, can be used to identify individuals that can benefit from modified treatment modalities. [0255] Antibodies can furthermore be useful for assessing expression of variant proteins in disease states, such as in active stages of a disease, or in an individual with a predisposition to a disease related to the function of the protein, in particular PD.

Antibodies specific for a variant protein of the present disclosure that is encoded by a nucleic acid that comprises at least one polymorphic marker or haplotype as described herein can be used to screen for the presence of the variant protein, for example to screen for a predisposition to PD as indicated by the presence of the variant protein.

[0256] Antibodies can be used in other methods. Thus, antibodies are useful as screening tools for evaluating proteins, such as variant proteins of the disclosure, in conjunction with analysis by electrophoretic mobility, isoelectric point, tryptic or other protease digest, or for use in other physical assays known to those skilled in the art.

Antibodies can also be used in tissue typing. In one such embodiment, a specific variant protein has been correlated with expression in a specific tissue type, and antibodies specific for the variant protein can then be used to identify the specific tissue type.

[0257] Subcellular localization of proteins, including variant proteins, can also be determined using antibodies, and can be applied to assess aberrant subcellular localization of the protein in cells in various tissues. Such use can be applied in genetic testing, but also in monitoring a particular treatment modality. In the case where treatment is aimed at correcting the expression level or presence of the variant protein or aberrant tissue distribution or, for instance, endometrial or blood cell expression of the variant protein, antibodies specific for the variant protein or fragments thereof can be used to monitor therapeutic efficacy.

[0258] Antibodies are further useful for inhibiting variant protein function, for example by blocking the binding of a variant protein to a binding molecule or partner. Such uses can also be applied in a therapeutic context in which treatment involves inhibiting a variant protein's function. An antibody can be for example be used to block or competitively inhibit binding, thereby modulating (i.e., agonizing or antagonizing) the activity of the protein. Antibodies can be prepared against specific protein fragments containing sites for specific function or against an intact protein that is associated with a cell or cell membrane.

[0259] The present disclosure also embodies the use of any pharmacologic agent that can be conjugated to an antibody or an antibody binding fragment, and delivered in active form. Examples of such agents include cytotoxins, radioisotopes, hormones such as a steroid, anti-metabolites such as cytosines, and chemotherapeutic agents. Other embodiments can include agents such as a coagulant, a cytokine, growth factor, bacterial endotoxin or a moiety of bacterial endotoxin. The targeting antibody-based agent directs the toxin to, and thereby selectively modulates the cell expressing the targeted surface receptor. In some embodiments, therapeutic antibodies employ cross-linkers that provide high in vivo stability (Thorpe et al., Cancer Res.. 48:6396 (1988)). In any event, it is proposed that agents such as these can, if desired, be successfully conjugated to antibodies or antibody binding fragments, in a manner that can allow their targeting, internalization, release or presentation at the site of the targeted cells expressing the EN associated biomarkers using known conjugation technology. For administration in vivo, for example, an antibody can be linked with an additional therapeutic payload, such as radionuclide, an enzyme, an immunogenic epitope, or a cytotoxic agent, including bacterial toxins (diphtheria or plant toxins, such as ricin). The in vivo half-life of an antibody or a fragment thereof can be increased by pegylation through conjugation to polyethylene glycol. Methods of Treatment

[0260] One embodiment of the present disclosure relates to methods of using compositions, e.g., pharmaceutical or neutraceutical compositions, and kits comprising agents that can reduce or increase the function and/or activity of polypeptides and/or nucleic acids that are associated with PD to inhibit or decrease PD progression, and/or are associated with complex I, II, III or IV, or lysosomal storage or metabolism. Another embodiment of the present disclosure provides methods, pharmaceutical or neutraceutical compositions, and kits for the treatment of animal subjects. The term "animal subject" as used herein includes humans as well as other mammals. The term "treating" as used herein . includes achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying cause of PD. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated PD such that an improvement is observed in the animal subject, notwithstanding the fact that the animal subject can still be afflicted with PD.

[0261] For embodiments where a prophylactic benefit is desired, a composition of the disclosure can be administered to a subject at risk of developing PD, or to a subject reporting one or more of the physiological symptoms of PD, even though a screening of the condition cannot have been made. Administration can prevent PD from developing, or it can reduce, lessen, shorten and/or otherwise ameliorate the progression of PD, or symptoms that develop. The pharmaceutical or neutraceutical composition can modulate a target PD associated biomarker. Wherein, the term modulate includes inhibition of PD associated biomarkers, complex I, II, III or IV, or lysosomal storage or metabolism associated genes, or alternatively activation of PD associated biomarkers or complex I, II, III or IV, or lysosomal storage or metabolism associated genes.

[0262] Reducing the activity and/or function of polypeptides and/or nucleic acids found to be associated with PD, and/or are associated with complex I, II, III or IV, or lysosomal storage or metabolism is also referred to as "inhibiting" the polypeptides and/or nucleic acids. The term "inhibits" and its grammatical conjugations, such as "inhibitory," do not require complete inhibition, but refer to a reduction in PD associated biomarkers' activities. In some embodiments, such reduction is by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, at least 90%, and can be by at least 95% of the activity of the enzyme in the absence of the inhibitory effect, e.g., in the absence of an inhibitor. Conversely, the phrase "does not inhibit" and its grammatical conjugations refer to situations where there is less than 20%, less than 10%, and can be less than 5%, of reduction in enzyme activity in the presence of the agent. Further the phrase "does not substantially inhibit" and its grammatical conjugations refer to situations where there is less than 30%, less than 20%, and in some embodiments less than 10% of reduction in enzyme activity in the presence of the agent.

[0263] Increasing the activity and/or function of polypeptides and/or nucleic acids found to be associated with PD, and/or are associated with complex I, II, III or IV, or lysosomal storage or metabolism is also referred to as "activating" the polypeptides and/or nucleic acids. The term "activated" and its grammatical conjugations, such as "activating," do not require complete activation, but refer to an increase in PD associated biomarkers' activities. In some embodiments such increase is by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and can be by at least 95% of the activity of the enzyme in the absence of the activation effect, e.g., in the absence of an activator.

Conversely, the phrase "does not activate" and its grammatical conjugations refer to situations where there can be less than 20%, less than 10%, and less than 5%, of an increase in enzyme activity in the presence of the agent. Further the phrase "does not substantially activate" and its grammatical conjugations refer to situations where there is less than 30%, less than 20%, and in some embodiments less than 10% of an increase in enzyme activity in the presence of the agent.

[0264] The ability to reduce enzyme activity is a measure of the potency or the activity of an agent, or combination of agents, towards or against the enzyme. Potency can be measured by cell free, whole cell and/or in vivo assays in terms of IC50, Ki and/or ED50 values. An IC50 value represents the concentration of an agent to inhibit enzyme activity by half (50%) under a given set of conditions. A Ki value represents the equilibrium affinity constant for the binding of an inhibiting agent to the enzyme. An ED50 value represents the dose of an agent to affect a half-maximal response in a biological assay. Further details of these measures will be appreciated by those of ordinary skill in the art, and can be found in standard texts on biochemistry, enzymology, and the like.

[0265] The present disclosure also includes kits that can be used to treat PD or

ET. These kits comprise an agent or combination of agents that inhibits PD or ET, inhibits a PD or ET associated biomarker and/or modulates complex I, II, III or IV, or lysosomal storage or metabolism associated genes or regions, and in some embodiments instructions teaching the use of the kit according to the various methods and approaches described herein. Such kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the agent. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like.

Kits, Arrays and Panels

[0266] Kits, arrays or panesl useful in the methods of the disclosure comprise components useful in any of the methods described herein, including for example, primers for nucleic acid amplification, hybridization probes for detecting genetic variation, or other marker detection, restriction enzymes, nucleic acid probes, optionally labeled with suitable labels, allele-specific oligonucleotides, antibodies that bind to an altered polypeptide encoded by a nucleic acid of the disclosure as described herein or to a wild type polypeptide encoded by a nucleic acid of the disclosure as described herein, means for amplification of genetic variations or fragments thereof, means for analyzing the nucleic acid sequence of nucleic acids comprising genetic variations as described herein, means for analyzing the amino acid sequence of a polypeptide encoded by a genetic variation, or a nucleic acid associated with a genetic variation, etc. The kits can for example include necessary buffers, nucleic acid primers for amplifying nucleic acids, and reagents for allele- specific detection of the fragments amplified using such primers and necessary enzymes (e.g., DNA polymerase). Additionally, kits can provide reagents for assays to be used in combination with the methods of the present disclosure, for example reagents for use with other screening assays for PD or ET.

[0267] In some embodiments, the disclosure pertains to a kit, array or panel for assaying a sample from a subject to detect the presence of a genetic variation, wherein the kit comprises reagents necessary for selectively detecting at least one particular genetic variation in the genome of the individual. In some embodiments, the disclosure pertains to a kit for assaying a sample from a subject to detect the presence of at least particular allele of at least one polymorphism associated with a genetic variation in the genome of the subject. In some embodiments, the reagents comprise at least one contiguous oligonucleotide that hybridizes to a fragment of the genome of the individual comprising at least genetic variation. In some embodiments, the reagents comprise at least one pair of oligonucleotides that hybridize to opposite strands of a genomic segment obtained from a subject, wherein each oligonucleotide primer pair is designed to selectively amplify a fragment of the genome of the individual that includes at least one genetic variation, or a fragment of a genetic variation. Such oligonucleotides or nucleic acids can be designed using the methods described herein. In some embodiments, the kit comprises one or more labeled nucleic acids capable of allele-specific detection of one or more specific polymorphic markers or haplotypes with ' a genetic variation, and reagents for detection of the label. In some embodiments, a kit, array or panel for detecting SNP markers can comprise a detection oligonucleotide probe, that hybridizes to a segment of template DNA containing a SNP polymorphisms to be detected, an enhancer oligonucleotide probe, detection probe, primer and/or an endonuclease, for example as described by Kutyavin et al. (Nucleic Acid Res., 34:el28 (2006)).

[0268] In some embodiments, the DNA template is amplified by any means of the present disclosure, prior to assessment for the presence of specific genetic variations as described herein. Standard methods well known to the skilled person for performing these methods can be utilized, and are within scope of the disclosure. In one such embodiment, reagents for performing these methods can be included in the reagent kit.

[0269] In a further aspect of the present disclosure, a pharmaceutical or neutraceutical pack (kit) is provided, the pack comprising a therapeutic agent and a set of instructions for administration of the therapeutic agent to humans screened for one or more variants of the present disclosure, as disclosed herein. The therapeutic agent can be a small molecule drug, an antibody, a peptide, an antisense or RNAi molecule, or other therapeutic molecules as described herein. In some embodiments, an individual identified as a carrier of at least one variant of the present disclosure is instructed to take a prescribed dose of the therapeutic agent. In one such embodiment, an individual identified as a carrier of at least one variant of the present disclosure is instructed to take a prescribed dose of the therapeutic agent. In some embodiments, an individual identified as a non-carrier of at least one variant of the present disclosure is instructed to take a prescribed dose of the therapeutic agent.

[0270] Also provided herein are articles of manufacture, comprising a probe that hybridizes with a region of human chromosome as described herein and can be used to detect a polymorphism described herein. For example, any of the probes for detecting polymorphisms described herein can be combined with packaging material to generate articles of manufacture or kits, arrays or panels. The kit can include one or more other elements including: instructions for use; and other reagents such as a label or an agent useful for attaching a label to the probe. Instructions for use can include instructions for screening applications of the probe for making a diagnosis, prognosis, or theranosis to PD or ET in a method described herein. Other instructions can include instructions for attaching a label to the probe, instructions for performing in situ analysis with the probe, and/or instructions for obtaining a sample to be analyzed from a subject. In some cases, the kit can include a labeled probe that hybridizes to a region of human chromosome as described herein.

[0271] The kit, array or panel can also include one or more additional reference or control probes that hybridize to the same chromosome or another chromosome or portion thereof that can have an abnormality associated with a particular endophenotype. A kit that includes additional probes can further include labels, e.g., one or more of the same or different labels for the probes. In other embodiments, the additional probe or probes provided with the kit can be a labeled probe or probes. When the kit further includes one or more additional probe or probes, the kit can further provide instructions for the use of the additional probe or probes. Kits for use in self-testing can also be provided. Such test kits . can include devices and instructions that a subject can use to obtain a biological sample (e.g., buccal cells, blood) without the aid of a health care provider. For example, buccal cells can be obtained using a buccal swab or brush, or using mouthwash.

[0272] Kits as provided herein can also include a mailer (e.g., a postage paid envelope or mailing pack) that can be used to return the sample for analysis, e.g., to a laboratory. The kit can include one or more containers for the sample, or the sample can be in a standard blood collection vial. The kit can also include one or more of an informed consent form, a test requisition form, and instructions on how to use the kit in a method described herein. Methods for using kits, arrays or panels are also included herein. One or more of the forms (e.g., the test requisition form) and the container holding the sample can be coded, for example, with a bar code for identifying the subject who provided the sample.

[0273] In some embodiments, an in vitro screening test can comprise one or more devices, tools, and equipment configured to collect a genetic sample from an individual. In some embodiments of an in vitro screening test, tools to collect a genetic sample can include one or more of a swab, a scalpel, a syringe, a scraper, a container, and other devices and reagents designed to facilitate the collection, storage, and transport of a genetic sample. In some embodiments, an in vitro screening test can include reagents or solutions for collecting, stabilizing, storing, and processing a genetic sample.

[0274] Such reagents and solutions for nucleotide collecting, stabilizing, storing, and processing are well known by those of skill in the art and can be indicated by specific methods used by an in vitro screening test as described herein. In some embodiments, an in vitro screening test as disclosed herein, can comprise a microarray apparatus and reagents, a flow cell apparatus and reagents, a multiplex nucleotide sequencer and reagents, and additional hardware and software necessary to assay a genetic sample for certain genetic markers and to detect and visualize certain genetic markers. [0275] The present disclosure further relates to kits, arrays or panels for using antibodies in the methods described herein. This includes, but is not limited to, kits, arrays or panels for detecting the presence of a variant protein in a test sample. One embodiment comprises antibodies such as a labeled or labelable antibody and a compound or agent for detecting variant proteins in a biological sample, means for determining tl^amount or the presence and/or absence of variant protein in the sample, and means for comparing the amount of variant protein in the sample with a standard, as well as instructions for use of the kit. In certain embodiments, the kit further comprises a set of instructions for using the reagents comprising the kit.

[0276] Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The following references contain embodiments of the methods and compositions that can be used herein: The Merck Manual of Diagnosis and Therapy, 18th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-18-2); Benjamin Lewin, Genes IX, published by Jones & Bartlett Publishing, 2007 (ISBN-13:

9780763740634); Kendrew et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1 -56081 -569-8).

[0277] Standard procedures of the present disclosure are described, e.g., in

Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1982); Sambrook et al., Molecular Cloning: A Laboratory Manual (2 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1989); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1986); or Methods in Enzymology: Guide to Molecular Cloning Techniques, Vol. 152, S. L. Berger and A. R. Kimmerl (eds.), Academic Press Inc., San Diego, USA (1987)). Current Protocols in Molecular Biology (CPMB) (Fred M. Ausubel, et al. ed., John Wiley and Sons, Inc.), Current Protocols in Protein Science (CPPS) (John E. Coligan, et. al., ed., John Wiley and Sons, Inc.), Current Protocols in Immunology (CPI) (John E. Coligan, et. al., ed. John Wiley and Sons, Inc.), Current Protocols in Cell Biology (CPCB) (Juan S. Bonifacino et. al. ed., John Wiley and Sons, Inc.), Culture of Animal Cells: A Manual of Basic Technique by R. Ian Freshney, Publisher: Wiley-Liss; 5th edition (2005), and Animal Cell Culture Methods (Methods in Cell Biology, Vol. 57, Jennie P. Mather and David Barnes editors, Academic Press, 1 st edition, 1998), which are all incorporated by reference herein in their entireties.

[0278] It should be understood that the following examples should not be construed as being limiting to the particular methodology, protocols, and compositions, etc., described herein and, as such, can vary. The following terms used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the embodiments disclosed herein.

[0279] Disclosed herein are molecules, materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of methods and compositions disclosed herein. It is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed and while specific reference of each various individual and collective combinations and permutation of these molecules and compounds cannot be explicitly disclosed, each is specifically contemplated and described herein. For example, if a nucleotide or nucleic acid is disclosed and discussed and a number of modifications that can be made to a number of molecules including the nucleotide or nucleic acid are discussed, each and every combination and permutation of nucleotide or nucleic acid and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed molecules and compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods, and that each such combination is specifically contemplated and should be considered disclosed.

[0280] Those skilled in the art can recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the method and compositions described herein. Such equivalents are intended to be encompassed by the following claims.

[0281] It is understood that the disclosed methods and compositions are not limited to the particular methodology, protocols, and reagents described as these can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present disclosure which can be limited only by the appended claims.

[0282] Unless defined otherwise, all technical and scientific terms used herein have the meanings that would be commonly understood by one of skill in the art in the context of the present specification.

[0283] It should be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "a nucleotide" includes a plurality of such nucleotides; reference to "the nucleotide" is a reference to one or more nucleotides and equivalents thereof known to those skilled in the art, and so forth. [0284] The term "and/or" shall in the present context be understood to indicate that either or both of the items connected by it are involved. While some embodiments of the present disclosure have been shown and described herein, it can be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions can now occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments of the disclosure described herein can be employed in practicing the disclosure. It is intended that the following claims define the scope of the disclosure and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Formulations, Routes of Administration, and Effective Doses

[0285] Yet another aspect of the present disclosure relates to formulations, routes of administration and effective doses for compositions comprising an agent or combination of agents of the instant disclosure. Such compositions can be used to treat a ND progression and a ND associated symptoms as described above.

[0286] Compounds of the disclosure can be administered as formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, transdermal patch, pulmonary, vaginal, suppository, or parenteral (including intramuscular, intraarterial, intrathecal, intradermal, intraperitoneal, subcutaneous and intravenous) administration or in a form suitable for administration by aerosolization, inhalation or insufflation. General information on drug delivery systems can be found in Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott Williams & Wilkins, Baltimore d. (1999).

[0287] In various embodiments, the composition includes carriers and excipients (including but not limited to buffers, carbohydrates, mannitol, proteins, polypeptides or amino acids such as glycine, bacteriostats, chelating agents, suspending agents, thickening agents and/or preservatives), water, oils including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, saline solutions, aqueous dextrose and glycerol solutions, flavoring agents, coloring agents, detackifiers and other acceptable additives, adjuvants, or binders, other pharmaceutically acceptable auxiliary substances to approximate physiological conditions, such as pH buffering agents, tonicity adjusting agents, emulsifying agents, wetting agents and the like. Examples of excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. In some embodiments, the pharmaceutical or neutraceutical preparation is substantially free of preservatives. In other embodiments, the pharmaceutical or neutraceutical preparation can contain at least one preservative. General methodology on pharmaceutical dosage forms is found in Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott, Williams, & Wilkins, Baltimore Md. (1999)). It can be recognized that, while any suitable carrier known to those of ordinary skill in the art can be employed to administer the compositions of this disclosure, the type of carrier can vary depending on the mode of administration.

[0288] Compounds can also be encapsulated within liposomes using well-known technology. Biodegradable microspheres can also be employed as carriers for the pharmaceutical or neutraceutical compositions of this disclosure. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268, 5,075,109,

5,928,647, 5,811,128, 5,820,883, 5,853,763, 5,814,344 and 5,942,252. .

[0289] The compound can be administered in liposomes or microspheres (or microparticles). Methods for preparing liposomes and microspheres for administration to a subject are well known to those of skill in the art. U.S. Pat. No. 4,789,734, the contents of which are hereby incorporated by reference, describes methods for encapsulating biological materials in liposomes. Essentially, the material is dissolved in an aqueous solution, the appropriate phospholipids and lipids added, and optionally along with surfactants, and the material dialyzed or sonicated, as necessary. A review of known methods is provided by G. Gregoriadis, Chapter 14, "Liposomes," Drug Carriers in Biology and Medicine, pp. 2. sup.87- 341 (Academic Press, 1979).

[0290] Microspheres formed of polymers or proteins are well known to those skilled in the art, and can be tailored for passage through the gastrointestinal tract directly into the blood stream. Alternatively, the compound can be incorporated and the microspheres, or composite of microspheres, implanted for slow release over a period of time ranging from days to months. See, for example, U.S. Pat. Nos. 4,906,474, 4,925,673 and 3,625,214, and Jein, TIPS 19:155-157 (1998), the contents of which are hereby incorporated by reference.

[0291] The concentration of drug can be adjusted, the pH of the solution buffered and the isotonicity adjusted to be compatible with intravenous injection, as is well known in the art.

[0292] The compounds of the disclosure can be formulated as a sterile solution or suspension, in suitable vehicles, well known in the art. The compositions can be sterilized by conventional, well-known sterilization techniques, or can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. Suitable formulations and additional carriers are described in Remington "The Science and Practice of Pharmacy" (20th Ed., Lippincott Williams & Wilkins, Baltimore MD), the teachings of which are incorporated by reference in their entirety herein. [0293] The agents or their pharmaceutically acceptable salts can be provided alone or in combination with one or more other agents or with one or more other forms. For example a formulation can comprise one or more agents in particular proportions, depending on the relative potencies of each agent and the intended indication. For example, in compositions for targeting two different host targets, and where potencies are similar, about a 1 :1 ratio of agents can be used. The two forms can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, aerosol spray, or packet of powder to be dissolved in a beverage; or each form can be formulated in a separate unit, e.g., two creams, two suppositories, two tablets, two capsules, a tablet and a liquid for dissolving the tablet, two aerosol sprays, or a packet of powder and a liquid for dissolving the powder, etc.

[0294] The term "pharmaceutically acceptable salt" means those salts which retain the biological effectiveness and properties of the agents used in the present disclosure, and which are not biologically or otherwise undesirable. For example, a pharmaceutically acceptable salt does not interfere with the beneficial effect of an agent of the disclosure in inhibiting a ND associated biomarkers' components

[0295] Typical salts are those of the inorganic ions, such as, for example, sodium, potassium, calcium, magnesium ions, and the like. Such salts include salts with inorganic or organic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, methanesulfonic acid, p toluenesulfonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, mandelic acid, malic acid, citric acid, tartaric acid or maleic acid. In addition, if the agent(s) contain a carboxy group or other acidic group, it can be converted into a pharmaceutically acceptable addition salt with inorganic or organic bases. Examples of suitable bases include sodium hydroxide, potassium hydroxide, ammonia,

cyclohexylamine, dicyclohexyl-amine, ethanolamine, diethanolamine, triethanolamine, and the like.

[0296] A pharmaceutically acceptable ester or amide refers to those which retain biological effectiveness and properties of the agents used in the present disclosure, and which are not biologically or otherwise undesirable. For example, the ester or amide does not interfere with the beneficial effect of an agent of the disclosure in inhibiting a ND associated biomarkers' components. Typical esters include ethyl, methyl, isobutyl, ethylene glycol, and the like. Typical amides include unsubstituted amides, alkyl amides, dialkyl amides, and the like.

[0297] In some embodiments, an agent can be administered in combination with one or more other compounds, forms, and/or agents, e.g., as described above.

Pharmaceutical or neutraceutical compositions comprising combinations of a ND associated biomarkers' inhibitors with one or more other active agents can be formulated to comprise certain molar ratios. For example, molar ratios of about 99:1 to about 1 :99 of a ND associated biomarkers' inhibitors to the other active agent can be used. In some subset of the embodiments, the range of molar ratios of ND associated biomarkers' inhibitors: other active agents are selected from about 80:20 to about 20:80; about 75:25 to about 25:75, about 70:30 to about 30:70, about 66:33 to about 33:66, about 60:40 to about 40:60; about 50:50; and about 90:10 to about 10:90. The molar ratio of ND associated biomarkers' inhibitors: other active agents can be about 1 :9, and in some embodiments can be about 1 :1 . The two agents, forms and/or compounds can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, or packet of powder to be dissolved in a beverage; or each agent, form, and/or compound can be formulated in separate units, e.g., two creams, suppositories, tablets, two capsules, a tablet and a liquid for dissolving the tablet, an aerosol spray a packet of powder and a liquid for dissolving the powder, etc.

[0298] If necessary or desirable, the agents and/or combinations of agents can be administered with still other agents. The choice of agents that can be co-administered with the agents and/or combinations of agents of the instant disclosure can depend, at least in part, on the condition being treated.

[0299] The agent(s) (or pharmaceutically acceptable salts, esters or amides thereof) can be administered per se or in the form of a pharmaceutical or neutraceutical composition wherein the active agent(s) is in an admixture or mixture with one or more pharmaceutically acceptable carriers. A pharmaceutical or neutraceutical composition, as used herein, can be any composition prepared for administration to a subject.

Pharmaceutical or neutraceutical compositions for use in accordance with the present disclosure can be formulated in conventional manner using one or more physiologically acceptable carriers, comprising excipients, diluents, and/or auxiliaries, e.g., which facilitate processing of the active agents into preparations that can be administered. Proper formulation can depend at least in part upon the route of administration chosen. The agent(s) useful in the present disclosure, or pharmaceutically acceptable salts, esters, or amides thereof, can be delivered to a subject using a number of routes or modes of administration, including oral, buccal, topical, rectal, transdermal, transmucosal, subcutaneous, intravenous, and intramuscular applications, as well as by inhalation.

[0300] For oral administration, the agents can be formulated readily by combining the active agent(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the agents of the disclosure to be formulated as tablets, including chewable tablets, pills, dragees, capsules, lozenges, hard candy, liquids, gels, syrups, slurries, powders, suspensions, elixirs, wafers, and the like, for oral ingestion by a subject to be treated. Such formulations can comprise pharmaceutically acceptable carriers including solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents. A solid carrier can be one or more substances which can also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. In powders, the carrier generally is a finely divided solid which is a mixture with the finely divided active component. In tablets, the active component generally is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted in the shape and size desired. The powders and tablets may contain from about one (1 ) to about seventy (70) percent of the active compound. Suitable carriers include but are not limited to magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. Generally, the agents of the disclosure can be included at concentration levels ranging from about 0.5%, about 5%, about 10%, about 20%, or about 30% to about 50%, about 60%, about 70%, about 80% or about 90% by weight of the total composition of oral dosage forms, in an amount sufficient to provide a desired unit of dosage.

[0301] Aqueous suspensions for oral use can contain agent(s) of this disclosure with pharmaceutically acceptable excipients, such as a suspending agent (e.g., methyl cellulose), a wetting agent (e.g., lecithin, lysolecithin and/or a long-chain fatty alcohol), as well as coloring agents, preservatives, flavoring agents, and the like.

[0302] In some embodiments, oils or non-aqueous solvents can be used to bring the agents into solution, due to, for example, the presence of large lipophilic moieties. Alternatively, emulsions, suspensions, or other preparations, for example, liposomal preparations, can be used. With respect to liposomal preparations, any known methods for preparing liposomes for treatment of a condition can be used. See, for example, Bangham et al., J. ol. Biol. 23: 238 (1965) and Szoka et al., Proc. Natl Acad. Sci. USA 75: 4194

(1978), incorporated herein by reference. Ligands can also be attached to the liposomes to direct these compositions to particular sites of action. Agents of this disclosure can also be integrated into foodstuffs, e.g., cream cheese, butter, salad dressing, or ice cream to facilitate solubilization, administration, and/or compliance in certain subject populations.

[0303] Pharmaceutical or neutraceutical preparations for oral use can be obtained

• as a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; flavoring elements, cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone (PVP). If desired, disintegrating agents can be added, such as the cross linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. The agents can also be formulated as a sustained release preparation.

[0304] Dragee cores can be provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents.

[0305] Pharmaceutical or neutraceutical preparations that can be used orally include push fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active agents can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for administration.

[0306] Other forms suitable for oral administration include liquid form preparations including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions, or solid form preparations which are intended to be converted shortly before use to liquid form preparations. Emulsions can be prepared in solutions, for example, in aqueous propylene glycol solutions or can contain emulsifying agents, for example, such as lecithin, sorbitan monooleate, or acacia. Aqueous solutions can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents. Aqueous suspensions can be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well known suspending agents. Suitable fillers or carriers with which the compositions can be administered include agar, alcohol, fats, lactose, starch, cellulose derivatives, polysaccharides, polyvinylpyrrolidone, silica, sterile saline and the like, or mixtures thereof used in suitable amounts. Solid form preparations include solutions, suspensions, and emulsions, and can contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.

[0307] A syrup or suspension can be made by adding the active compound to a concentrated, aqueous solution of a sugar, e.g., sucrose, to which can also be added any accessory ingredients. Such accessory ingredients can include flavoring, an agent to retard crystallization of the sugar or an agent to increase the solubility of any other ingredient, e.g., as a polyhydric alcohol, for example, glycerol or sorbitol. [0308] When formulating compounds of the disclosure for oral administration, it can be desirable to utilize gastroretentive formulations to enhance absorption from the gastrointestinal (Gl) tract. A formulation which is retained in the stomach for several hours can release compounds of the disclosure slowly and provide a sustained release that can be in some embodiments of the disclosure. Disclosure of such gastro-retentive formulations are found in Klausner, E.A.; Lavy, E.; Barta, M.; Cserepes, E.; Friedman, M. ; Hoffman, A., Pharm. Res., 20:1466 (2003), Hoffman, A.; Stepensky, D.; Lavy, E.; Eyal, S. Klausner, E.; Friedman, M., Int. J. Pharm.. H:141 (2004), Streubel, A.; Siepmann, J.; Bodmeier, R.: Expert Qpin. Drug Deliver., 3:217 (2006), and Chavanpatil, .D.; Jain, P.; Chaudhari, S.; Shear, R.; Vavia, P.R., Int. J. Pharm. (2006). Expandable, floating and bioadhesive techniques can be utilized to maximize absorption of the compounds of the disclosure.

[0309] The compounds of the disclosure can be formulated for parenteral administration (e.g., by injection, for example bolus injection or continuous infusion) and can be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example solutions in aqueous polyethylene glycol.

[0310] For injectable formulations, the vehicle can be chosen from those known in art to be suitable, including aqueous solutions or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles. The formulation can also comprise polymer compositions which are biocompatible, biodegradable, such as poly(lactic-co-glycolic)acid. These materials can be made into micro or nanospheres, loaded with drug and further coated or derivatized to provide superior sustained release performance. Vehicles suitable for periocular or intraocular injection include, for example, suspensions of therapeutic agent in injection grade water, liposomes and vehicles suitable for lipophilic substances. Other vehicles for periocular or intraocular injection are well known in the art.

[0311] In an embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical or neutraceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition can also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.

[0312] When administration is by injection, the active compound can be formulated in aqueous solutions, specifically in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer. The solution can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active compound can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In some embodiments, the pharmaceutical or

' neutraceutical composition does not comprise an adjuvant or any other substance added to enhance the immune response stimulated by the peptide. In some embodiments, the pharmaceutical or neutraceutical composition comprises a substance that inhibits an immune response to the peptide. Methods of formulation are known in the art, for example, as disclosed in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Co., Easton P.

[0313] In addition to the formulations described previously, the agents can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation or transcutaneous delivery (for example subcutaneously or intramuscularly), intramuscular injection or use of a transdermal patch. Thus, for example, the agents can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

[0314] In some embodiments, pharmaceutical or neutraceutical compositions comprising one or more agents of the present disclosure exert local and regional effects when administered topically or injected at or near particular sites of infection. Direct topical application, e.g., of a viscous liquid, . solution, suspension, dimethylsulfoxide (DMSO)-based solutions, liposomal formulations, gel, jelly, cream, lotion, ointment, suppository, foam, or aerosol spray, can be used for local administration, to produce for example local and/or regional effects. Pharmaceutically appropriate vehicles for such formulation include, for example, lower aliphatic alcohols, polyglycols (e.g., glycerol or polyethylene glycol), esters of fatty acids, oils, fats, silicones, and the like. Such preparations can also include preservatives (e.g., p-hydroxybenzoic acid esters) and/or antioxidants (e.g., ascorbic acid and tocopherol). See also Dermatological Formulations: Percutaneous absorption, Barry (Ed.), Marcel Dekker Incl, 1983.

[0315] Pharmaceutical or neutraceutical compositions of the present disclosure can contain a cosmetically or dermatologically acceptable carrier. Such carriers are compatible with skin, nails, mucous membranes, tissues and/or hair, and can include any conventionally used cosmetic or dermatological carrier within this context. Such carriers can be readily selected by one of ordinary skill in the art. In formulating skin ointments, an agent or combination of agents of the instant disclosure can be formulated in an oleaginous hydrocarbon base, an anhydrous absorption base, a water-in-oil absorption base, an oil-in- water water-removable base and/or a water-soluble base. Examples of such carriers and excipients include, but are not limited to, humectants (e.g., urea), glycols (e.g., propylene glycol), alcohols (e.g., ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), pyrrolidones, glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.

[0316] Ointments and creams can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions can be formulated with an aqueous or oily base and can in general also containing one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001 ,139. Such patches can be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.

[0317] Lubricants which can be used to form pharmaceutical or neutraceutical compositions and dosage forms of the disclosure include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof. Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof. A lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical or neutraceutical composition.

[0318] The compositions according to the present disclosure can be in any form suitable for topical application, including aqueous, aqueous-alcoholic or oily solutions, lotion or serum dispersions, aqueous, anhydrous or oily gels, emulsions obtained by dispersion of a fatty phase in an aqueous phase (O/W or oil in water) or, conversely, (W/O or water in oil), microemulsions or alternatively microcapsules, microparticles or lipid vesicle dispersions of ionic and/or nonionic type. These compositions can be prepared according to conventional methods. Other than the agents of the disclosure, the amounts of the various constituents of the compositions according to the disclosure are those conventionally used in the art. These compositions in particular constitute protection, treatment or care creams, milks, lotions, gels or foams for the face, for the hands, for the body and/or for the mucous membranes, or for cleansing the skin. The compositions can also consist of solid preparations constituting soaps or cleansing bars.

[0319] Compositions of the present disclosure can also contain adjuvants common to the cosmetic and dermatological fields, such as hydrophilic or lipophilic gelling agents, hydrophilic or lipophilic active agents, preserving agents, antioxidants, solvents, fragrances, fillers, sunscreens, odor-absorbers and dyestuffs. The amounts of these various adjuvants are those conventionally used in the fields considered and, for example, are from about 0.01 % to about 20% of the total weight of the composition. Depending on their nature, these adjuvants can be introduced into the fatty phase, into the aqueous phase and/or into the lipid vesicles.

[0320] In some embodiments, ocular viral infections can be effectively treated with ophthalmic solutions, suspensions, ointments or inserts comprising an agent or combination of agents of the present disclosure. Eye drops can be prepared by dissolving the active ingredient in a sterile aqueous solution such as physiological saline, buffering solution, etc., or by combining powder compositions to be dissolved before use. Other vehicles can be chosen, as is known in the art, including but not limited to: balance salt solution, saline solution, water soluble polyethers such as polyethyene glycol, polyvinyls, such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate. If desired, additives ordinarily used in the eye drops can be added. Such ' additives include isotonizing agents (e.g., sodium chloride, etc.), buffer agent (e.g., boric acid, sodium monohydrogen phosphate, sodium dihydrogen phosphate, etc.), preservatives (e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, etc.), thickeners (e.g., saccharide such as lactose, mannitol, maltose, etc.; e.g., hyaluronic acid or its salt such as sodium hyaluronate, potassium hyaluronate, etc.; e.g., mucopolysaccharide such as chondroitin sulfate, etc.; e.g., sodium polyacrylate, carboxyvinyl polymer, crosslinked polyacrylate, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose or other agents known to those skilled in the art).

[0321] The solubility of the components of the present compositions can be enhanced by a surfactant or other appropriate co-solvent in the composition. Such cosolvents include polysorbate 20, 60, and 80, Pluronic F68, F-84 and P-103, cyclodextrin, or other agents known to those skilled in the art. Such co-solvents can be employed at a level of from about 0.01 % to 2% by weight.

[0322] The compositions of the disclosure can be packaged in multidose form.

Preservatives can be preferred to prevent microbial contamination during use. Suitable preservatives include: benzalkonium chloride, thimerosal, chlorobutanol, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, Onamer M, or other agents known to those skilled in the art. In the prior art ophthalmic products, such preservatives can be employed at a level of from 0.004% to 0.02%. In the compositions of the present application the preservative, e.g., benzalkonium chloride, can be employed at a level of from 0.001 % to less than 0.01 %, e.g. from 0.001 % to 0.008%, e.g., about 0.005% by weight. It has been found that a concentration of benzalkonium chloride of 0.005% can be sufficient to preserve the compositions of the present disclosure from microbial attack.

[0323] In some embodiments, ND associated symptoms of the ear can be effectively treated with otic solutions, suspensions, ointments or inserts comprising an agent or combination of agents of the present disclosure.

[0324] In some embodiments, the agents of the present disclosure are delivered in soluble rather than suspension form, which allows for more rapid and quantitative absorption to the sites of action. In general, formulations such as jellies, creams, lotions, suppositories and ointments can provide an area with more extended exposure to the agents of the present disclosure, while formulations in solution, e.g., sprays, provide more immediate, short-term exposure.

[0325] In some embodiments relating to topical/local application, the

pharmaceutical or neutraceutical compositions can include one or more penetration enhancers. For example, the formulations can comprise suitable solid or gel phase carriers or excipients that increase penetration or help delivery of agents or combinations of agents of the disclosure across a permeability barrier, e.g., the skin. Many of these penetration- enhancing compounds are known in the art of topical formulation, and include, e.g., water, alcohols (e.g., terpenes like methanol, ethanol, 2-propanol), sulfoxides (e.g., dimethyl sulfoxide, decylmethyl sulfoxide, tetradecylmethyl sulfoxide), pyrrolidones (e.g., 2- pyrrolidone, N-methyl-2-pyrrolidone, N-(2-hydroxyethyl)pyrrolidone), laurocapram, acetone, dimethylacetamide, dimethylformamide, tetrahydrofurfuryl alcohol, L-a-amino acids, anionic, cationic, amphoteric or nonionic surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), fatty acids, fatty alcohols (e.g., oleic acid), amines, amides, clofibric acid amides, hexamethylene lauramide, proteolytic enzymes, a-bisabolol, d-limonene, urea and N,N- diethyl-m-toluamide, and the like. Additional examples include humectants (e.g., urea), glycols (e.g., propylene glycol and polyethylene glycol), glycerol monolaurate, alkanes, alkanols, ORGELASE, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and/or other polymers. In some embodiments, the pharmaceutical or neutraceutical compositions can include one or more such penetration enhancers.

[0326] In some embodiments, the pharmaceutical or neutraceutical compositions for local/topical application can include one or more antimicrobial preservatives such as quaternary ammonium compounds, organic mercurials, p-hydroxy benzoates, aromatic alcohols, chlorobutanol, and the like.

'[0327] Gastrointestinal ND symptoms can be effectively treated with orally- or rectally delivered solutions, suspensions, ointments, enemas and/or suppositories comprising an agent or combination of agents of the present disclosure.

[0328] Respiratory ND symptoms can be effectively treated with aerosol solutions, suspensions or dry powders comprising an agent or combination of agents of the present disclosure. Administration by inhalation is particularly useful in treating viral infections of the lung, such as influenza. The aerosol can be administered through the respiratory system or nasal passages. For example, one skilled in the art can recognize that a composition of the present disclosure can be suspended or dissolved in an appropriate carrier, e.g., a pharmaceutically acceptable propellant, and administered directly into the lungs using a nasal spray or inhalant. For example, an aerosol formulation comprising a ND associated biomarkers' inhibitors can be dissolved, suspended or emulsified in a propellant or a mixture of solvent and propellant, e.g., for administration as a nasal spray or inhalant. Aerosol formulations can contain any acceptable propellant under pressure, such as a cosmetically or dermatologically or pharmaceutically acceptable propellant, as conventionally used in the art.

[0329] ' An aerosol formulation for nasal administration is generally an aqueous solution designed to be administered to the nasal passages in drops or sprays. Nasal solutions can be similar to nasal secretions in that they are generally isotonic and slightly buffered to maintain a pH of about 5.5 to about 6.5, although pH values outside of this range can additionally be used. Antimicrobial agents or preservatives can also be included in the formulation.

[0330] An aerosol formulation for inhalations and inhalants can be designed so that the agent or combination of agents of the present disclosure is carried into the respiratory tree of the subject when administered by the nasal or oral respiratory route. Inhalation solutions can be administered, for example, by a nebulizer. Inhalations or insufflations, comprising finely powdered or liquid drugs, can be delivered to the respiratory system as a pharmaceutical or neutraceutical aerosol of a solution or suspension of the agent or combination of agents in a propellant, e.g. , to aid in disbursement. Propellants can be liquefied gases, including halocarbons, for example, fluorocarbons such as fluorinated chlorinated hydrocarbons, hydrochlorofluorocarbons, and hydrochlorocarbons, as well as hydrocarbons and hydrocarbon ethers.

[0331] Halocarbon propellants useful in the present disclosure include fluorocarbon propellants in which all hydrogens are replaced with fluorine,

chlorofluorocarbon propellants in which all hydrogens are replaced with chlorine and at least one fluorine, hydrogen-containing fluorocarbon propellants, and hydrogen-containing chlorofluorocarbon propellants. Halocarbon propellants are described in Johnson, U.S. Pat. No. 5,376,359; Byron et al., U.S. Pat. No. 5,190,029; and Purewal et al., U.S. Pat. No. 5,776,434. Hydrocarbon propellants useful in the disclosure include, for example, propane, isobutane, n-butane, pentane, isopentane and neopentane. A blend of hydrocarbons can also be used as a propellant. Ether propellants include, for example, dimethyl ether as well as the ethers. An aerosol formulation of the disclosure can also comprise more than one propellant. For example, the aerosol formulation can comprise more than one propellant from the same class, such as two or more fluorocarbons; or more than one, more than two, more than three propellants from different classes, such as a fluorohydrocarbon and a hydrocarbon. Pharmaceutical or neutraceutical compositions of the present disclosure can also be dispensed with a compressed gas, e.g., an inert gas such as carbon dioxide, nitrous oxide or nitrogen.

[0332] Aerosol formulations can also include other components, for example, ethanol, isopropanol, propylene glycol, as well as surfactants or other components such as oils and detergents. These components can serve to stabilize the formulation and/or lubricate valve components.

[0333] The aerosol formulation can be packaged under pressure and can be formulated as an aerosol using solution ' s, suspensions, emulsions, powders and semisolid preparations. For example, a solution aerosol formulation can comprise a solution of an agent of the disclosure such as a ND associated biomarkers' inhibitors in (substantially) pure propellant or as a mixture of propellant and solvent. The solvent can be used to dissolve the agent and/or retard the evaporation of the propellant. Solvents useful in the disclosure include, for example, water, ethanol and glycols. Any combination of suitable solvents can be use, optionally combined with preservatives, antioxidants, and/or other aerosol components.

[0334] An aerosol formulation can also be a dispersion or suspension. A suspension aerosol formulation can comprise a suspension of an agent or combination of agents of the instant disclosure, e.g., a ND associated biomarkers' inhibitors, and a dispersing agent. Dispersing agents useful in the disclosure include, for example, sorbitan trioleate, oleyl alcohol, oleic acid, lecithin and corn oil. A suspension aerosol formulation can also include lubricants, preservatives, antioxidant, and/or other aerosol components. •[0335] An aerosol formulation can similarly be formulated as an emulsion. An emulsion aerosol formulation can include, for example, an alcohol such as ethanol, a surfactant, water and a propellant, as well as an agent or combination of agents of the disclosure, e.g., a ND associated biomarkers' inhibitors.. The surfactant used can be nonionic, anionic or cationic. One example of an emulsion aerosol formulation comprises, for example, ethanol, surfactant, water and propellant. Another example of an emulsion aerosol formulation comprises, for example, vegetable oil, glyceryl monostearate and propane.

[0336] The compounds of the disclosure can be formulated for administration as suppositories. A low melting wax, such as a mixture of triglycerides, fatty acid glycerides, Witepsol S55 (trademark of Dynamite Nobel Chemical, Germany), or cocoa butter is first melted and the active component is dispersed homogeneously, for example, by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and to solidify.

[0337] The compounds of the disclosure can be formulated for vaginal administration. Pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.

[0338] It is envisioned additionally, that the compounds of the disclosure can be attached releasably to biocompatible polymers for use in sustained release formulations on, in or attached to inserts for topical, intraocular, periocular, or systemic administration. The controlled release from a biocompatible polymer can be utilized with a water soluble polymer to form an instillable formulation, as well. The controlled release from a biocompatible polymer, such as for example, PLGA microspheres or nanospheres, can be utilized in a formulation suitable for intra ocular implantation or injection for sustained release administration, as well any suitable biodegradable and biocompatible polymer can be used.

[0339] In one aspect of the disclosure, the subject's carrier status of any of the genetic variation risk variants described herein, or genetic variants identified via other analysis methods within the genes or regulatory loci that are identified by the CNVs described herein, can be used to help determine whether a particular treatment modality for a ND, such as any one of the above, or a combination thereof, should be administered. The present disclosure also relates to methods of monitoring progress or effectiveness of a treatment option for a ND. The treatment option can include any of the above mentioned treatment options commonly used. This can be done based on the outcome of determination of the presence of a particular genetic variation risk variant in the individual, or by monitoring expression of genes that are associated with the variants of the present disclosure. Expression levels and/or mRNA levels can thus be determined before and during treatment to monitor its effectiveness. Alternatively, or concomitantly, the status with respect to a genetic variation, and or genotype and/or haplotype status of at least one risk variant for a ND presented herein can determined before and during treatment to monitor its effectiveness.

[0340] Alternatively, biological networks or metabolic pathways related to the genes within, or associated with, the genetic variations described herein can be monitored by determining mRNA and/or polypeptide levels. This can be done for example, by monitoring expression levels or polypeptides for several genes belonging to the network and/or pathway, in samples taken before and during treatment. Alternatively, metabolites belonging to the biological network or metabolic pathway can be determined before and during treatment. Effectiveness of the treatment is determined by comparing observed changes in expression levels/metabolite levels during treatment to corresponding data from healthy subjects.

[0341] In a further aspect, the genetic variations described herein and/or those subsequently found (e.g., via other genetic analysis methods such as sequencing) via targeted analysis of those genes initially identified by the genetic variations described herein, can be used to increase power and effectiveness of clinical trials. Thus, individuals who are carriers of at least one at-risk genetic variation can be more likely to respond to a particular treatment modality for a ND. In some embodiments, individuals who carry at-risk variants for gene(s) in a pathway and/or metabolic network for which a particular treatment is targeting are more likely to be responders to the treatment. In some embodiments, individuals who carry at-risk variants for a gene, which expression and/or function is altered by the at-risk variant, are more likely to be responders to a treatment modality targeting that gene, its expression or its gene product. This application can improve the safety of clinical trials, but can also enhance the chance that a clinical trial can demonstrate statistically significant efficacy, which can be limited to a certain sub-group of the population. Thus, one possible outcome of such a trial is that carriers of certain genetic variants are statistically significant and likely to show positive response to the therapeutic agent. Further, one or more of the genetic variations employed during clinical trials for a given therapeutic agent can be used in a companion diagnostic test that is administered to the patient prior to administration of the therapeutic agent to determine if the patient is likely to have favorable response to the therapeutic agent.

[0342] In a further aspect, the genetic variations described herein can be used for targeting the selection of pharmaceutical or neutraceutical agents for specific individuals. The agent can be any of the agents described in the above. Personalized selection of treatment modalities, lifestyle changes or combination of the two, can be realized by the utilization of the at-risk genetic variations or surrogate markers in linkage disequilibrium with the genetic variations. Thus, the knowledge of an individual's status for particular genetic variations can be useful for selection of treatment options, for example for treatments that target genes or gene products affected by one or more of the genetic variations. Certain combinations of variants, including those described herein, but also combinations with other risk variants for a ND, can be suitable for one selection of treatment options, while other variant combinations can target other treatment options. Such combinations of variants can include one variant, two variants, three variants, or four or more variants, as needed to determine with clinically reliable accuracy the selection of treatment module.

[0343] The invention will be further described by the following non-limiting examples.

Example 1

[0344] Sanger sequencing was performed on all 477 cases in the PD cohort.

Exons and flanking sequence of the PD candidate gene NUBPL were sequenced bi- directionally. Briefly, PCR amplification was carried out in an 5 μΙ amplification solution comprising AmpliTaq Gold.RTM, PCR Master Mix (Applied Biosystems), a solution containing the target polynucleotide, and a forward PCR primer and reverse PCR primer (as indicated below).

[0345] The PCR samples were thermal cycled to conduct PCR in a thermal cycler. A two-step "boost/nest" PCR strategy was used. An initial boost reaction generating a larger fragment was performed, followed by a nest reaction, using the initial product as a template for the nest. The nest product was then sequenced. All products were sequenced on ABI 3730XL DNA sequencers.

[0346] Millipore Montage PCR384 plates were used for PCR cleanup (the boost reaction was not cleaned up, only the nest reaction). The primers utilized were as follows:

[0347] In Table 5, the primers can be described as follows: BST 5' and BST 3' are the boost primers, 5' and 3' respectively; NST 5' and 3' are the nest primers; B-LEN and N-LEN are the lengths of the boost and nest products.

[0348] Sequencing of the DNA was performed asd follows: A 5 microliter reaction volume was thermocycled using an Eppendorf Mastercycler 384 according to the following program: (a) 1 minute hold at 96°C, (b) 25 cycles of 10 seconds at 96°C, then 5 seconds at 50°C, followed by 60°C for 4 minutes. The samples were then held at 4°C. BigDye 3.1 chemistry was used for sequencing. Millipore SEQ384 plates were used for dye terminator removal.

[0349] Known and novel variants (SNPs/SNVs/indels) were identified and interpreted using NCBI's dbSNP, the Exome Variant Server (EVS) database hosted by a website at the University of Washington (evs.gs.washington.edu/EVS/), or the Exome Aggregation Consortium (ExAC) database hosted by a website at the Broad Institute (http://exac.broadinstitute.org/) to assess their frequency in the general population. NUBPL was selected for Sanger sequencing on the basis of its high odds ratio - (OR) and strong links to PD relevant biology. It is impacted by CNVs in 15 PD cases (2 familial and 13 idiopathic). Assessment (via PubMed and OMIM) of NUBPL's gene function revealed a direct link to mitochondrial dysfunction (Calvo et al. 2010), specifically complex I deficiency, a well-known phenotype in PD patients (Schapira et al. 1989; Schapira 1993). However, Complex I deficiency (OMIM 252010) is a mitochondrial disorder (often occurring in newborns) considered to be distinct from PD and NUBPL mutations have never been reported in PD patients. All 10 exons of NUBPL in 477 PD patients were sequenced. The majority of sequencing variants (SNVs or small indels) were found at greater than ~1 % frequency in dbSNP, the EVS database, or the ExAC database and thus assumed to be benign. Some NUBPL variants were found in these databases at low frequency (<1 % frequency) or were novel (not present in the databases) and these may be. rare, benign variants or are potentially causative of disease. For example, pathogenic mutations that cause autosomal recessive disorders will be found in the general or unselected population in a heterozygous state.

Pathogenic mutations can also be found in general or unselected populations in homozygous (autosomal recessive), heterozygous (autosomal dominant), or compound heterozygous (autosomal recessive) states wherein such individuals have milder symptoms of the disorder and remain undiagnosed, or may develop the disease at a later age (e.g. , Alzheimer's disease or Parkinsons' disease, which typical manifest as late onset disorders). NUBPL variants (CNVs, indel, and SNVs) were found in the PD cohort of 467 cases (CGH experiments passing quality control) or 477 cases (Sanger sequencing experiments on full cohort). Three SNVs were novel (never reported in dbSNP, EVS exome, ExAC exome databases) and found to be significantly associated with PD (p-value <0.05). One is a synonymous variant [c.120C>G; p. (A40=)],and two are intronic variants adjacent to exons (13 or 18 nucleotides from the slice site).

Example 2

[0350] Mitochondrial dysfunction has been repeatedly associated with Parkinson's disease (PD). While four redox complexes (I - IV) comprise the electron transport chain in mitochondria, reduced complex I (CI) activity is the most common mitochondrial defect and is found in PD (Mounsey et al., Parkin. Pis., 61:7472 (2011); Schapira et al., Lancet. 1:1269 (1989)). Despite the biochemical evidence for impaired CI activity, no nuclear CI genes have been associated with PD. While there have been reports associating genetic variants in mitochondrial DNA and PD, these have been inconsistent, and sometimes conflicting (Hudsone et al., Neurology, 80:2042 (2013); Coskun et al., BBA, 1820:553 (2012)). By contrast, CI deficiency (MIM 252010), a rare heterogeneous disorder that often manifests in children with failure to thrive, developmental delay, and lactic acidosis, is known to occur via an autosomal recessive mechanism. To date, mutations in 23 nuclear-encoded CI genes and 6 mitochondrial- encoded CI genes have been described as causative of CI deficiency (Fassone & Rahman, J. Med. Genet., 49:578 (2011 )).

[0351] Genetic findings, such as those shown in Table 6, support a potential association between PD and the CI gene nucleotide binding protein-like {NUBPL, gene aliases IND1 , hulndl , C14orf127). Since the identification of NUBPL as a CI assembly factor in 2008 (Bych et al., EMBO J.. 27:1736 (2008); Sheftel et al., Mol. Cell Biol.. 29:6059 (2009)) and a 2010 report on a pediatric patient with CI deficiency with both alleles of NUBPL impacted by pathogenic mutations (Calvo et al., Nat. Genet., 42:851 (2010)), NUBPL mutations have been reported in six additional unrelated cases with CI deficiency (Kevelam et al., Neuroloav.80:1577 (2013) ; Tenish et al., Neurology, 79:391 (2012)). Interestingly, all patients carrying mutations in the NUBPL gene also had signs of leukoencephalopathy characterized by MRI patterns with specific abnormalities in the cerebellar cortex and subcortical white matter (Kevelem et al., 2013). Furthermore, three new cases of CI deficiency are reported herein (Table 6, cases 8 and 9, wherein case 8 corresponds to a family with two affected siblings).

[0352] NUBPL was identified as a potential PD gene in a genome-wide screen for copy number variants (CNVs). In a study of 467 PD cases, one patient was found with a complex chromosomal rearrangement that disrupted NUBPL and was identical to the one found in the first reported case of CI deficiency caused by NUBPL (Calvo et al., 2010; Tucker et al., Hum. Mutant. 33:411 (2012)). Exon sequencing of our PD cohort revealed additional known and novel sequence variants in the NUBPL gene associated with PD that, along with the chromosomal rearrangement, provide the first evidence that heterozygous carriers of NUBPL mutations may be at risk for developing PD.

Materials and Methods

PD Cohort [0353] Genomic DNA samples from blood and clinical data from 477 PD patients undergoing clinical care at the Parkinson's Institute comprise the PD cohort used in the genetic studies herein. The demographics of the cohort are the following: familial (27%) and sporadic (73%) cases, a greater number of male (65%) vs. female (35%) patients, and both early-onset (<50 years of age, 10%) and late-onset cases (>50 years of age; 90%) with a median age of 67 years. Patients with known mutations in PD genes SNCA (PARK 1/4), LRRK2 (PARK8), PARK2 (Parkin), PINK1 (PARK6), PARK7 (DJ- 1), and GBA were excluded from this study. All subjects have been clinically assessed by movement disorder specialists with a neurological history and physical examination that includes standardized diagnostic rating by Gelb criteria (Gelb et al., Arch. Neurol.. 56:33 (1999)). An Institutional Review Board approved the study and all participants provided informed consent.

Control Cohort

[0354] For control samples, a cohort biobanked at Population Diagnostics, Inc. (Melville, NY) was used, which is comprised of genomic DNA samples derived from blood. Briefly, the cohort is comprised of 1 ,005 reportedly healthy donors of European ancestry (505 males and 500 females) greater than 45 years of age. Further details on this control cohort are provided elsewhere (Prasad et al., G3 (Bethesda), 2:1665 (2012)). Donors were consented and de-identified via a protocol approved by the Institutional Review Board. Genome-wide CNV data on this control cohort were used to interpret the CNV data generated on the PD cohort.

Genome-wide copy number variant (CNV) analysis

[0355] CNV detection on DNA samples from the control and PD cohorts was performed on commercially available comparative genomic hybridization microarrays containing 1 million probes (1 M CGH array) (catalog design #021529; Agilent

Technologies, Santa Clara, CA). The experiments were performed using a 2-color labeling and hybridization format wherein the control or PD DNA sample is labeled with Cy3 and the Reference DNA is labeled with Cy5, followed by co-hybridization of the two labeled samples to the microarray. All experiments were performed with the same, sex- matched Reference DNA (healthy male or female donor, genomic DNA was isolated from whole blood). Raw data were generated in an ISO-certified service laboratory (Oxford Gene Technology, Oxford, UK) and array images and feature-extracted data files were archived and further processed at Population Diagnostics, Inc. UK, Oxford, UK using the CNV-calling algorithm DNAcopy to generate the CNV calls [14]. Quality control (QC) metrics that assesses sample and array data quality were applied to the PD experiments, with 98% of the 477 PD samples passing QC (yielding 467 PD experiments for CNV analysis).

NUBPL chromosomal rearrangement breakpoint sequencing

[0356] . The chromosomal rearrangement identified in patient PI- 1256 was validated using the polymerase chain reaction (PCR) primers described in Prasad et al. (2012) to confirm it was identical to the one found in a pediatric case with CI deficiency (Calvo et al. (2010); Prasad et al. (2012)).

NUBPL exon sequencing

[0357] Sanger sequencing was performed on all 477 cases in the PD cohort. Exons and flanking sequence of NUBPL were sequenced bi-directionally by

Polymorphic DNA Technologies Inc. (Alameda, CA). Known and novel NUBPL variants in Table 6 were identified and interpreted using control data from NCBI's dbSNP, the 1000 Genomes Project (Genomes Project C, Nature. 491 :56 (2012)), the Exome Variant Server, NHLBI GO Exome Sequencing Project (ESP) database [URL:

http://evs.gs.washington.edu/EVS/], and the Exome Aggregation Consortium (ExAC) database [URL: http://exac.broadinstitute.org/].

Table 6. Comparison of NUBPL rare and pathogenic variants and clinical summary for CI deficiency and PD cases.

NUBPL splice site prediction

[0358] Splice site prediction was performed on intronic variants and the synonymous variant (p.A40=) in Table 6 using the Human Splicing Finder (HSF, v.2.4.1 ), which is hosted at: http://www.umd.be/HSF/ (Desmet et al., Nucl. Acids Res.. 37:e67 (2009)). Splice site analysis for the synonymous variant predicted potentially altered splicing via three mechanisms: activation of an exonic cryptic donor site, destruction of an exonic splicing enhancer (ESE) motif, and creation of an exonic splicing silencer (ESS) motif. Splice site analysis for the six intronic variants predicted altered splicing via creation or destruction of ESE or ESS motifs, alteration of the splice donor and/or acceptor site, or alteration of the branch point.

Results

Genome-wide CNV analysis identifies a PD patient with a chromosomal rearrangement impacting NUBPL

[0359] The genome-wide CNV screen identified one PD patient with a complex chromosomal rearrangement on chromosome 14 that disrupts the NUBPL gene (Figures 1 A, B) via deleted (254 Kb) and duplicated (134 Kb) regions. The

rearrangement was not found in 1 ,005 controls and PCR analysis (Figure 1 B) and breakpoint sequencing confirmed that this rearrangement is identical to the previously described alteration in a child with CI deficiency (Calvo et al., 2010; Tucker et al., 2012). Sequencing of NUBPL identifies three novel NUBPL variants in the PD cohort

[0360] Three unique SNVs were identified in the NUBPL gene that were statistically significant (FET <0.05), were not reported in unselected (i.e., 'control') populations (dbSNP, l OOOGenomes, EVS, and ExAC databases), and had odds ratios (ORs) of 75.93 - 207.16: a synonymous SNV [c.120C>G; p.(A40=)] and two intronic

SNVs (c.694-18A>T and c.815-13T>C). Three additional known SNVs were found in the PD cohort at higher frequency relative to controls (ORs 1.82-21.14), but did not reach significance, although c.693+7G>A and c. 897+49T>G were nominally associated (FET 0.06-0.08). The c.-1 C>T variant is potentially protective against PD, with an OR value of 0.34 (FET = 0.0086). The remaining SNVs were found at appreciable frequency in the SNV databases and are likely benign (ORs 0.45-1 .06), with the exception of c.815- 27T>C, as discussed below.

[0361] Three PD cases in the cohort were found to have a c.815-27T>C aberrant splicing mutation that was previously found in all 7 unrelated CI deficiency cases (Calvo et al., 2010; Kevelem et al., 2013; Tucker et al., 2012; Tenisch et al. 2012). However, the PD cases did not carry a second non-synonymous c.166G>A (p.G56R) variant that was present on the same allele in all CI deficiency patients with the c.815-27T>C mutation. Thus, the three PD cases have an alternate haplotype than is found in the majority of CI deficiency patients. The frequency of the two haplotypes (c.815-27T>C found in this PD cohort and c.815-27T>C plus c.166G>A found in CI deficiency patients) is presently unknown in the general population but the frequency of each variant, independent of haplotype, in the ExAC db of about 33,000 European (non- Finnish) ancestry subjects is 0.98% for the c.815-27T>C variant and 0.054% for the c.166G>A variant, which indicates that the CI deficiency patients have a rarer haplotype as was found in the present PD cases.

In silico analysis of functional impact of NUBPL variants

[0362] The novel synonymous SNV [c.120C>G; p.(A40=)] corresponding to an alanine at position 40 in the NUBPL protein may be pathogenic (Hunt et al., Methods Mol. Biol., 578:23 (2009); Kimchi-Sarfaty et al., Science, 315:525 (2007) ; Sauna et al., Nat. Rev. Genet. , 12:683 (2011 )). There is ample evidence that these variants can be causative by producing splicing defects, altering RNA stability/structure, or impacting protein translation rates (Sauna et al., 2011). Analysis of the c.120C>G [p.(A40=)] variant revealed that the GCC codon (wild-type) is more frequently used than the GCG codon (novel in this PD cohort), a change from 27.7 to 7.4, which is one of highest differentials found for the 20 amino acids (Nakamura et al., Nucl. Acids Res., 28:292 (2000)). While this change in codon frequency suggests a potential mechanism of pathogenicity, functional validation will be required.

Clinical summary for patients with PD-associated NUBPL variants

[0363] Clinically, all patients had typical late-onset PD (3 familial, 6 sporadic) with an age of onset range of 60-73 years. Most patients had a good response to levodopa except for the patient with the complex rearrangement of the NUBPL gene, who presented with a more rapidly progressing form of PD and autonomic involvement. See Table 7 for an overview of the patient's clinical features.

[0364] Interestingly, newly reported cases of CI deficiency (Table 6, cases 8 and 9) and newly ascertained family history on a previously reported case (Table 6, case 4) report a family history of ET, PD, or RLS in the maternal and paternal lines for these cases, whichs further supports the association of NUBPL variants with PD. In fact, thus far, all families with one or more patients diagnosed with NUBPL Complex I deficiency report a history of movement disorders in older family members (family histories could not be ascertained for 6 of 9 families, cases 1 -3 and 5-7 in Table 6).

[0365] Of further note there are certain clinical symptoms that have been reported in PD patients and/or their family members, which are known to be (or are likely) carriers of NUBPL mutations or variants, including but not limited to tremor, which is a symptom in both PD and essential tremor (ET), and restless legs syndrome (RLS). For example, tremor was reported in the mother of PD patient ID 1870, who is reported to have the NUBPL variant c.815-13T>C. Furthermore, two PD patient (IDs 1256 and 1801 ) with NUBPL variants were diagnosed with RLS and the sister {NUBPL variant status unknown, DNA was unavailable) of patient ID 1256 was also diagnosed with RLS. Overlapping phenotypes and co-occurrence of RLS and PD have been reported (e.g., see Peeraully and Tan 2012, PMID 23211049) but more studies are needed, especially if a correlation exists only for PD cases with specific genetic subtypes. For example, the genetic subtype of PD may consist of clinical features more consistent with multiple system atrophy (MSA). Evidence for this was found for the clinical presentation in PD patient ID 1256 (who has a chromosomal rearrangement that disrupts NUBPL), which was compatible with diagnosis of PD, but differential diagnosis of multiple system atrophy (MSA) was also considered because of the presence of urinary incontinence, low blood pressure, and absence of clear benefit from L-dopa (e.g., see Stamelou et al. 2013, PMID 23720239; Fereshtehnejad and Lokk, PMID 24634790). Given the constellation of movement disorder symptoms and overlap between neurological diseases such as PD, ET, RLS, and MSA, it can be appreciated by those skilled in the art that co-occurence of two or more clinical diagnoses (e.g., PD and ET, PD and RLS, or ET and RLS) is not unexpected and in fact has been reported in the literature (e.g., see Peeraully and Tan 2012, PMID 23211049; Zimprich 2011, PMID 21734494; Vilarino-Guell et al. 2010, PMID 20369371 ; Raiput et al. 2014, PMID 25118025). The molecular pathology of neurological disorders can also be distinct or overlapping. For example, high or low irons levels can be a hallmark feature of a particular disorder, such as high brain iron levels in PD patients and iron deficiency in RLS patients, yet there are many reported cases of PD co-occurring with RLS. Thus, it can be appreciated by those skilled in the art that, depending on the specific molecular (e.g. , decreased complex I activity) or genetic (e.g., pathogenic NUBPL mutations) subtype of the disorder, a tailored treatment regimen may involve use of opposing therapeutic strategies such as iron supplementation vs. iron chelation (e.g., see Satija and Ondo 2008, PMID 1848792; Hare et al. 2013, PMID 23874300; Ayton et al. 2014, PMID 25011704) depending on which molecular or genetic subtype is found in the patient. In other words, some diagnosed cases of PD (and/or RLS, ET, MSA, etc.) may benefit from iron supplementation, whereas other cases may benefit from iron chelation therapy. Genetic and molecular evidence have been found that pathogenic or associated variants in NUBPL, an Fe-S protein required for proper assembly of complex I, link the neurological disorders complex I deficiency, PD, ET, and RLS, which is consistent with the genotype-phenotype correlations reported herein (Table 6). Furthermore, genetic evidence for the role of NUBPL in RLS is corroborated by report of a linkage peak on chromosome 14 that was mapped in an Italian kindred (see Bonati et al. PMID 12764067). While the Bonati et al. 2003 study did not pinpoint a specific gene in the linkage interval, the region is immediately adjacent to the NUBPL locus, which is a complex repetitive region in the genome. In fact, a second linkage peak reported in Bonati et al. 2003 does encompass the NUBPL gene, which lies between microsatellite markers D14S275 and D14S70. It can be appreciated by those skilled in the art that other genes involved in mitochondrial dysfunction, particularly variants found in other complex I genes, including but not limited to NDUFAF2, NDUFC2, NDUFC2-KCTD14, NDUFS4, and NDUFV1 , may have associations not only with PD, but also ET, RLS, and MSA, or any combination of symptoms described for this set of neurological disorders.

[0366]

Discussion

[0367] This study provides the first genetic evidence of an association between

PD and the NUBPL gene, which had previously been reported to cause CI deficiency in seven pediatric cases via a recessive mechanism (Calvo et al. Nat Genet. 2010 Oct;42(10):851 -8; Kevelam et al. Neurology. 2013 Apr 23;80(17):1577-83; Tenisch et al. Neurology. 2012 Jul 24;79(4):391 ; Tucker et al. Hum Mutat. 2012 Feb;33(2) :411-8). Precedents exist for genes causing early onset and severe clinical presentation when both alleles contain pathogenic mutations, and milder symptoms and later onset when only one allele is impacted by a deleterious mutation. As with the established association between PD and the GBA gene that causes the rare disorder Gaucher disease (MIM 606463) (Sidransky et al. N Engl J Med. 2009 Oct 22;361 (17):1651 -61 ; Sidransky Discov Med. 2012 Oct;14(77):273-81 ), it can be appreciated by those skilled in the art that individuals heterozygous for NUBPL mutations may have an increased risk for development of PD. Also reported herein are three new patients with CI deficiency in two unrelated families that are compound heterozygotes for known mutations and one novel mutation (p.L104P) and, for the first time, report the presence of ET, PD, RLS, or tremor in families with a CI deficiency patient. Indeed, out of three such families, for which family histories were available, all demonstrate this association (two new families described herein and one family from a previously reported case (Kevelam et al. 2013). These new genetic findings in PD and CI deficiency, along with the link to other movement disorders, add to the extensive evidence for mitochondrial dysfunction in early and late onset neurological disorders and provide a basis for the nomination of NUBPL as a new gene that causes or contributes to PD pathology, and ET and RLS.

[0368] The important findings are that one of the PD patients has an identical chromosomal rearrangement of the NUBPL gene as was found in a patient with CI deficiency and that three novel NUBPL SNVs (Table 6) in the PD cohort have an OR > 76 and are statistically significant. It is not surprising to discover a genetic association between PD and mitochondrial complex I gene NUBPL given that mitochondrial impairment is one of the major disease-associated mechanisms of neurodegeneration found in PD (reviewed in (Schapira et al., 1989; Henchcliffe and Beal, Nat. Clin. Pract. Neurol., 4:600 (2005)). Furthermore, several mitochondrial toxins (e.g., MPTP or rotenone) inhibit CI activity and cause nigrostriatal cell death and have been utilized extensively in vivo and in vitro to model PD.

[0369] The NUBPL protein is required for assembly of the CI enzyme

(NADH:ubiquinone oxidoreductase), which is the largest (about 1 MDa) of the respiratory chain components and is comprised of 45 subunits (38 are nuclear-encoded, 7 are mitochondrial-encoded (Scheftel et al., 2009). Nine assembly factors have recently been found to cause CI deficiency (Nouws et al., Brain, 135:12 (2012)) but NUBPL may be particularly important, because, thus far, it is the only CI assembly factor that is an Fe/S protein and it likely transfers Fe/S to Cl's 8 Fe/S clusters (Scheftel et al. , 2009), which is critical for the mitochondrial electron transport.

[0370] An interesting parallel to the present NUBPL genetic findings is the

GBA gene and the increased risk for heterozygous carriers developing PD. In 1996, the first report of GBA mutations in PD appeared (Neudorfer et al., QJM. 89:691 (1990)), followed by numerous papers with supporting but inconclusive results. It was not until a large multi-center study conducted with over 5,000 patients and controls that firmly established GBA mutations were associated with typical late-onset PD (Sidransky et al., NEJM, 36J.:1651 (2009)). Mutations in the GBA gene cause autosomal-recessive Gaucher's disease but a single mutation in the GBA gene is predisposing (5-fold increased risk) to late-onset typical PD (Sidransky, Discov. Med.. 14:273 (2012)).

[0371] An analogous mechanism for the NUBPL gene is hypothesized, wherein loss of function mutations impacting both alleles cause CI deficiency in children and young adults, while carriers of NUBPL pathogenic mutations have a higher risk for developing late-onset PD. Another similar example was recently described in three PD cases who are heterozygous carriers of NPC1 mutations, which causes Niemann-Pick disease (MIM 257220) (Kluenemann et al., J. Neuro. Sci.. 335:219 (2013)).

[0372] Given the overlap in NUBPL mutations between CI deficiency and PD, the compelling and well-established mitochondrial dysfunction as a key mechanism of PD, and analogous findings for an increased risk in heterozygous GBA mutation carriers for PD, broader screening of larger cohorts of cases and controls via sequencing and copy number microarrays is warranted to support the association between NUBPL variants and PD. Functional validation of PD-associated variants will further confirm if heterozygous carriers of NUBPL variants that reduce CI activity are at increased risk for PD.

Example 3

[0373] The data was generated on the basis of a comparison of copy number variants (CNVs) identified in 2 cohorts:

1. 1 ,005 Normal individuals (Normal Variation Engine - NVE);

2. 565 Parkinsons Disease (PD) cases (477 samples obtained from The

Parkinson's Institute and Clinical Center, Sunnyvale, CA 94085, USA and 87 samples obtained from the Coriell Institute, Camden, NJ, USA - NINDS cohort details can be found at

http://ccr.coriell.org/Sections/Collections/NINDS/DNAPanelDe tail.aspx7Pan elld=NDPT088&Pgld=436.)

Genomic DNA sample hybridization - NVE and PD cohorts

[0374] Genomic DNA samples from individuals within the Normal cohort (NVE

'test' subjects) and from the PD cohort (PD 'test' subjects) were hybridized against a single, sex-matched reference individual as follows. Reference DNA samples were labeled with Cy5 and test subject DNA samples were labeled with Cy3. After labeling, samples were combined and co-hybridized to Agilent 1 M feature oligonucleotide microarrays, design ID 021529 (Agilent Product Number G4447A) using standard conditions (array Comparative Genomic Hybridization - aCGH). Post-hybridization, arrays were scanned at 2 μιτι resolution, using Agilent's DNA microarray scanner, generating tiff images for later analysis.

[0375] All tiff images were analyzed using Agilent Feature Extraction (FE) software, with the following settings:

Human Genome Freeze: hg18:NCBI36:Mar2006

FE version: 10.7.3.1

Grid/design file: 021529_D_F_20091001

Protocol: CGH_107_Sep09

[0376] This procedure generates a variety of output files, one of which is a text-tab delimited file, containing about 1 ,000,000 rows of data, each corresponding to a specific feature on the array. This * .txt file was used to perform CNV calling using DNAcopy, an open source software package implemented in R via BioConductor (http://www.bioconductor.org/packages/release/bioc/html/DNAc opy.html). Losses or gains were determined according to a threshold log2ratio, which was set at -/+0.35. In other words, all losses with a Iog2ratio value <= -0.35 were counted, as were all gains with a log2ratio >= +0.35. All log2ratio values were determined according to Cy3/Cy5 (Test/Reference). A minimum probe threshold for CNV-calling was set at 2 (2 consecutive probes were sufficient to call a CNV). A CNV list was thus generated for each individual in the 2 cohorts.

[0377] There were a total of 162,316 CNVs in the NVE cohort of 1 ,005 individuals (an average of 162 CNVs per individual). Using custom scripts, these CNVs (many of which appeared in multiple individuals) were 'merged' into a master list (NVE- master) of non-redundant CNV-subregions, according to the presence or absence of the CNV-subregion in individuals within the cohort. Using this approach, the NVE-master list has 14,693 distinct CNV-subregions, some of which are uniquely present in a single individual and some of which are present in multiple individuals. For example, consider 3 individuals within the NVE cohort with the following hypothetical CNVs:

A. Chr1 : 1 -100,000;

B. Chr1 : 10,001 -100,000;

C. Chr1 :1 -89,999;

In the master list, these would be merged into 3 distinct CNV subregions, as follows: CNV-subregion 1 Chr1 :1 -10,000 Patients A, C

CNV-subregion 2 Chr1 :10,001 -89,999 Patients A, B, C

CNV-subregion 3 Chr90,000:1 -100,000 Patients A, B

[0378] There were a total of 88,627 CNVs in the PD cohort of 565 individuals

(an average of 157 CNVs per individual). Using custom scripts, these CNVs (many of which appeared in multiple individuals) were 'merged' into a master list (PD-master) of non-redundant CNV-subregions, according to the presence or absence of the CNV- subregion in individuals within the cohort. Using this approach, the PD-master list has 11,584 distinct CNV-subregions, some of which are uniquely present in a single individual and some of which are present in multiple individuals.

[0379] CNV-subregions of interest were obtained after:

1. Annotation using custom designed scripts in order to attach to each CNV region relevant information regarding overlap with known genes and exons;

2. A calculation of the odds ratio (OR) for each CNV-subregion, according to the following formula:

* OR = (PD/(100-PD))/(NVE/(1005-NVE))

where:

PD = number of PD individuals with CNV-subregion of interest

NVE = number of NVE individuals with CNV-subregion of interest

As an illustrative example, consider the CNV subregion chr2:99221389-99281387, which is found in 1 individual in the NVE cohort and 3 individuals in the PD cohort. The OR is:

(3/(562))/(1 /(1004)) = 5.35

Note that, by one convention, if either of NVE or PD = 0, a value of 0.5 is added to all 4 entries in the main formula above * , in order to avoid dealing with infinities. This has the effect of artificially lowering OR values in cases where no individuals within the NVE have the CNV. This method is applicable to all the calculations in Figures 8-11. This method is also used when calculating the Fisher's 2-tailed Exact Test (FET) in the event that any one of the variables is zero.

[0380] CNV-subregions/genes that fulfill one of the following criteria were identified:

1. Strong biology linking the CNV-subregion and/or the gene it overlaps, with known pathways/mechanisms or biology in PD (in some cases, statistical evidence is lacking but does not exclude the CNV-subregion as a candidate)

2. Statistical analysis combined with strong biology without obvious biological connection (best FET in this category was 3.25E-10);

It can be appreciated by those skilled in the art that the number of PD candidate CNV- subregions, irrespective of category, may increase or decrease as additional PD cohorts are analyzed.

Description of Sequence Data

[0381] The sequence file contains genomic sequence information for (in the following order):

A. All distinct CNVs listed in Figure 8A;

B. The full genomic extent of the transcripts listed in 11A;

Note that:

1. Higher priority SEQ IDs have lower numbers. Thus, SEQ ID NO:1 represents the highest priority gene, etc;

I 29 2. SEQ ID NOs:1 -197 are the CNV sequences from Figure 8A;

3. SEQ ID NOs:198-805 are the transcript sequences from Figure 11 A.

Examples of sequences:

SEQ ID NO:1 = 80,262 bp CNV (gain) at chr2:99201125-99281387 involving genes LYG1 , LYG2; and SEQ ID NO:198 = LYG2, transcript NM_175735, which is 12,860 bp in length:

[0382] The basis of this application with novel PD genes was to mine the PD cohort CNV data for:

1. Gains or losses impacting 1 -2 PD cases but are not found in Normals (n = 1 ,005) and that impact genes with strong PD-relevant biology.

2. Gains or losses occurring in intergenic regions but are near genes with strong PD-relevant biology.

Thus, this data mining process is biology-driven. Even a single PD case with a CNV can be causal or contributing to PD on the basis of the following criteria:

1. There are fewer CNVs in the genome relative to other classes of variants (e.g.,

SNVs); so one PD case with a CNV that is not found in Normals may be relevant.

2. CNVs, due to their large size (> about 5,000 bp), are more likely to impact the function or expression of a gene as compared to an SNV (1 bp).

3. Assessment of the known biology for the gene with respect to PD.

Additional genes, regiosn are included in Figures 8D, 9D, 10D and 11 D. Those include protective variants (PVs) as well as causal variants (CVs) Because PVs may be completely absent from PD cases, and, in order to define herein the extent and details of the CNVs that are protective, Figure 8B lists the original CNVs for all participants in the study (cases and controls). Hence, some of the CNV entries listed in Figure 8B are found only in controls. This is made clearer inFigure 9B, wherein the actual

CNV_subregions are detailed, including the multiple occurrences in NVE cases (normals) for those subregions not found in PD cases. An extra column has been added to Figure 9B, to delineate which CNV_subregions are being tagged as PVs (low ORs);

[0383] Figure 8 shows exemplary regions with genetic variations that are associated with PD. For each variation, the following may be provided: chromosome, original CNV start, original CNV stop, original CNV size, CNV type, PD case ID, RefSeq gene symbol, and SEQ ID No. corresponding to that region. Figures 8A-D list all CNVs of interest, with the exception that, for each entry, the original CNV start and stop positions are noted, along with original CNV size, type (loss or gain), case ID and gene annotation (for the CNV-subregion NOT original CNV). Figure 8A provides CNVs corresponding regions associated with SEQ ID NOs:1 -197. Figure 8B provides CNVs corresponding to regions in SEQ ID NOs:1059-1340. Figure 8C provides CNVs corresponding to regions in SEQ ID NOs:1621 -2002. Figure 8D provides CNVs corresponding to regions in SEQ ID NOs:806-916. The final column in Figure 8A contains SEQ ID numbers for exemplary genes/CNV subregions, which also correspond to higher priority genes/CNV subregions. Thus, SEQ ID NO:1 has the highest priority, SEQ ID NO:2 has the next highest priority.

[0384] Figure 9 shows exemplary subregions with genetic variations that are associated with PD. For each variation, the following may be provided: chromosome, CNV subregion start, CNV subregion stop, CNV subregion size, CNV type, PD case ID(s), RefSeq gene symbol, exon overlap, NVE cases, PD cases, FET, OR, and category. Figures 9A-D are similar to Figures 8A-D but there are a number of exceptions. Firstly, the CNV coordinates listed refer to the actual CNV-subregions found to be unique or significantly different between the disease and normal cohorts, as opposed to Figure 8, which lists the original CNVs. Secondly, an extra column details whether genie CNV-subregions of interest overlap an exon or not. Third and fourth, 2 extra columns detail the number of normal cases and the number of disease cases that harbor the relevant CNV-subregion. Finally, 3 columns report Fisher's 2-tailed Exact Test (FET), odds ratio (OR) and the Category under which the CNV-subregion falls wrt significance. Figure 9A provides CNV subregions corresponding to regions associated with SEQ ID NOs:1 -197. Figure 9B provides CNV subregions corresponding to SEQ ID NOs: 1059-1.340. Figure 9C provides CNV subregions corresponding to SEQ ID NOs: 1621 -2002. Figure 9D provides CNV subregions corresponding to regions associated with SEQ ID NOs:806-916

[0385] Figure 10 is a summary of the characteristics of the regions associated with PD. Figure 10A provides CNV subregions corresponding to regions associated with SEQ ID NOs:1 -197 in. Figure 11 B provides CNV subregions corresponding to SEQ ID NOs: 1059-1340. Figure 10C provides CNV subregions corresponding to SEQ ID NOs: 1621 -2002. Figure 10D provides CNV subregions corresponding to SEQ ID NOs:806-916

[0386] Figure 11 is a summary of transcripts in the regions associated with PD and SEQ ID numbers therefor. Figure 11 A is a summary of transcripts associated with SEQ ID NOs:1 -197, e.g., those having SEQ ID Nos. 198-805. Figure 11 B is a summary of transcripts associated with SEQ I D NOs: 1059-1340, e.g., those corresponding to SEQ ID Nos.1341 -1620. Figure 11C is a summary of transcripts associated with SEQ ID NOs:1621 -2002, e.g., those having SEQ ID NOs:2003-2640. Figure 11 D is a summary of transcripts associated with SEQ ID NOs:806-915, e.g., those having SEQ ID Nos. 918-1042.

[0387] An example that illustrates this process is NUBPL on the basis of statistics alone (a total of 15 cases were found to have a CNV: 1 case with a large 364Kb CNV + 14 cases with an identical small 2.6Kb CNV). However, if only the large NUBPL CNV had been considered, the biology-driven analysis algorithm would have found:

1. The large CNV found in 1 PD case is not found in 1 ,005 Normals (OR = 6.45). 2. The large CNV disrupts over half of the NUBPL gene, which would result in loss of function for this allele.

3. Assessment of the biology for NUBPL in the literature (PubMed) reveals that it is a cause of Complex I (CI) deficiency when both alleles are mutated

(autosomal recessive disorder). Decreased CI activity is a hallmark pathology of PD that has been known for many years and lower CI activity has been observed for some mutations even if only 1 allele is mutated.

Example 4

[0388] Figure 2 illustrates a 10 exonic deletion in NQ01. While NQ01 common variants have been linked to PD, reassessment of these studies

(http://www.pdgene.org/) found there was no association for most. The GWAS catalog of published studies does not list any NQ01 variants for PD or for any disease or phenotype. However, there is ample evidence that up-regulation of NQ01 is neuroprotective in PD and this 10 Kb loss of function rare variant is likely to be pathogenic.

[0389] Figure 3 shows a large triplication associated with lysozyme G-like genes. While limited gene information is available for the goose-type (G-like) genes, another lysozyme family member (LYZ) can form amyloid fibrils and LYZ mutations are know to cause Familial Amyloidosis (OMIM 153450). Gene triplication is a mutational mechanism that occurs for the known PD gene SNCA and, interestingly, 1 of the 3 PD cases with the LYG2 LYG1 triplication was suspected of having an SNCA triplication based on a brain pathology report but CGH analysis showed no copy number changes impacting the patient's SNCA gene.

[0390] Figure 4 depicts nonoverlapping deletions in SUMF1 . While the 210 Kb and 81 Kb deletions have partial overlap with CNVs found in normal subjects (see Normal Cohort annotation track, CNVs denoted by gray bars), there is a region of overlap for these two deletions with no CNVs found in normal subjects. Autosomal recessive mutations in SUMF1 are known to cause multiple sulfatase deficiency (OMIM 607939). Lysosomal defects associated with neurodegeneration are found in a mouse model with knockout of SUMF1 .

[0391] Figure 5 illustrates an intergenic deletion. The deletion is located about

6 Kb away from the 3' end of the RASSF3 and GNS genes, which is a region that contains transcription factor (TF) binding sites, such as PD-relevant TFs REST (aka NRSF) and YY1. Autosomal recessive mutations in GNS, a lysosomal pathway gene, are known to cause mucopolysaccharidosis type MID, also known as Sanfilippo syndrome type D (OMIM 607939).

[0392] Figure 6 is a schematic of PD genes in the lysosomal pathway.

Gaucher's disease is an autosomal recessive disorder caused by mutations in GBA with well-established evidence that individuals heterozygous for GBA mutations have an increased risk for PD. Thus, a similar pathogenic mechanism may occur for other genes in the lysosomal pathway: severe, early onset disease when both gene copies are impacted by loss of function mutations, but late onset PD when one gene copy is knocked out. Similarly, several other lysosomal pathway genes cause autosomal recessive Mucopolysaccharidosis (MPSVSanfilippo disorders and other lysosomal storage disorders and potentially increase the risk for PD via an autosomal dominant mechanism. Loss of function CNVs (or those that may result in decreased gene expression) are reported herein for ARSB, CERK, GALNS, GNS (intergenic) PSAP, SCARB1 , SCARB2, SMPD4, and SUMF1.

[0393] Figure 7 illustrates a protective variant. A 33 Kb intergenic duplication was detected in 0 PD cases and 15 normal subjects (subset of 6 are shown, OR = 0.07, FET = 0.005). The CNV is located in an intergenic region upstream of adjacent genes CITED4 and CTPS1 and impacts a strong transcription factor binding region (ENCODE annotation tracks, UCSC genome browser). The expression level of CITED4 and/or CTPS1 may be altered due to presence or absence of this CNV. Recently, nucleotide metabolism has been implicated in a PINK1 fly model of PD and CTP synthase 1 (CTPS1, aka CTP synthetase 1 ) is a key enzyme that converts UTP to CTP. CTP synthase 1 deficiency (OMIM 123860) is an autosomal recessive immunodeficiency disorder and SNCA was recently found to play a role in hematopoiesis, B cell lymphopoiesis and adaptive immune response.

[0394] Figure 12 illustrates a protective variant. A 78-83 Kb deletion was detected in 0 PD cases and 9 normal subjects (8 of 9 subjects have an identical deletion and the ninth subject's deletion is substantially overlapped, OR = 0.11 , FET = 0.036).

The CNV impacts MSR1 (macrophage scavenger receptor 1 , aka SCARA 1 and SR-A).

The gene is linked to neuroinflammation and diabetes, both of which are increasingly implicated in pathological mechanisms of PD.

[0395] Other protective variants include:

RefSeq Gene RefSeq Gene

Symbol_extra Symbol

AGMO_intergenic AGMO

AK8 AK8

ATG7 ATG7

BASP1 BASP1

CCSER1 . CCSER1

CGNL1 CGNL1

CITED4_intergenic CITED4

CTNNA3 CTNNA3

CTPS1_intergenic CTPS1

DGKBjntergenic DGKB

DOCK4_intergenic DOCK4

FADDjntergenic FADD

GSTA2 GSTA2 IMMP2L_intergenic IMMP2L

IRX2 IRX2

IRX4 IRX4

ITSN2_intergenic ITSN2

KCNIP4_intergenic KCNIP4

LCN15_intergenic LCN 15

LOC401177 LOC401177

LOFMntergenic LOR

MSR1 MSR1

NCOA1_intergenic NCOA1

NR1 H4 NR1 H4

NTRK2_intergenic NTRK2

PARK2 PARK2

PLXNA4_intergenic PLXNA4

PPFIA1_intergenic PPFIA1

PPM1 L PPM1 L

PREPL PREPL

PRR9_intergenic PRR9

RBM47 RBM47

SLC28A3_intergenic SLC28A3

SLC3A1 SLC3A1

SPAG16 SPAG 16

ST3GAL4 ST3GAL4

SYNDIG1_intergenic SYNDIG1

TMEM141_intergenic TMEM141

TMEM2_intergenic TMEM2 .

TOP3B TOP3B

TRPM3_intergenic TRPM3

WDR72 WDR72

Example 5

[0396] A therapeutic approach for treating PD on the basis of a specific genetic subtype (i.e., presence of PD-associated variants within a specific gene) was assessed for PD genes. Two strategies were used to match therapies to genes:

1. Assess which PD genes are known drug targets (see, e.g.,Agarwal et al., Nat.

Rev. Drug Discov.12(8):575-6 (2013n.

2. Studies that support a therapeutic mechanism of action related to a specific PD gene on the basis of two subcategories: a) prescription-based therapies requiring approval by the FDA (or related bodies in other countries, such as the European Medicines Agency) that are already approved or are in clinical trials,

b) over-the-counter (OTC) therapies that do not require prescription.

[0397] Gene-specific therapeutic strategies are specified in Figure 7. Column

A lists the gene symbol (if the CNV is near a gene with PD-relevant biology, 'intergenic'is appended to the gene symbol). Column B indicates 'yes' if the gene is a known drug target according to the database described in Agarwal et al., supra. Column C lists specific types of therapies that require approval by government agencies. Column D lists specific types of therapies that are available over-the-counter (OTC), but also includes dietary actions.

[0398] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.