Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NEURAL-DERIVED HUMAN EXOSOMES FOR ALZHEIMER'S DISEASE AND CO-MORBIDITIES THEREOF
Document Type and Number:
WIPO Patent Application WO/2021/096929
Kind Code:
A1
Abstract:
Methods for using gene expression changes and mutations in neural organoids to identify neural networks that predict the onset of Alzheimer's disease and exosome RNA that can be used to predict onset and act as therapeutic targets are disclosed.

Inventors:
ANAND RENE (US)
MCKAY SUSAN (US)
Application Number:
PCT/US2020/059971
Publication Date:
May 20, 2021
Filing Date:
November 11, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ANAND RENE (US)
International Classes:
C12N5/078; A61K35/12; A61K35/14; A61K35/30; A61P25/28; C12N5/071; C12N5/074
Domestic Patent References:
WO2002103015A22002-12-27
WO2019200383A12019-10-17
Other References:
LU 'O'NG, KV ET AL.: "The role of thiamine in autism", AMERICAN JOURNAL OF PSYCHIATRY AND NEUROSCIENCE, vol. 1, no. 2, 2013, pages 22 - 37, XP055541227, DOI: 10.11648/j.ajpn. 20130102 .11
Attorney, Agent or Firm:
HELWIG, Bryan G. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for treating Alzheimer’s disease in a human, using a patient-specific pharmacotherapy, the method comprising: a) procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; d) collecting a biological sample from the patient specific neural organoid; e) detecting changes in Alzheimer's disease biomarker expression from the patient specific neural organoid sample that are differentially expressed in humans with Alzheimer’s disease;

0 performing assays on the patient specific neural organoid to identify therapeutic agents that alter the differentially expressed Alzheimer’s disease biomarkers in the patient-specific neural organoid sample; and g) administering a therapeutic agent for Alzheimer’s disease to treat the human.

2. The method of claim 1 , wherein the at least one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast derived from skin or blood cells from humans.

3. The method of claim 2, wherein the fibroblast derived skin or blood cells from humans is identified with the genes identified in Table 1 , Table 2, Table 5, or Table 7.

4. The method of claim 1 , wherein the measured biomarkers comprise nucleic acids, proteins, or their metabolites such as glucose.

5. The method of claim 1 , wherein the measured biomarkers comprise on or a plurality of biomarkers identified in Table 1 , Table 2, Table 5 or Table 7 or variants thereof and can be correlated with disease onset, severity, or progression.

6. The method of claim 5, further wherein a combination of biomarkers is detected, the combination comprising a nucleic acid encoding human A2M, APR variants and one or a plurality of biomarkers comprising a nucleic acid encoding human genes identified in Table

1. 7. The method of claim 1 , wherein the neural organoid biological sample is collected after about one hour up to about 12 weeks post inducement.

8. The method of claim 7, wherein the neural organoid sample is procured from structures of the neural organoid that mimic structures developed in utem at about 5 weeks.

9. The method of claim 7, wherein the neural organoid at about twelve weeks postinducement comprises encoded structures and cell types of retina, cortex, midbrain, hindbrain, brain stem, or spinal cord.

10. The method of claim 7, wherein the neural organoid contains microglia, and one or a plurality of Alzheimer’s disease biomarkers as identified in Table 1 and Table 7.

11. The method of claim 1 , wherein the method is used to detect environmental factors that cause or exacerbate Alzheimer’s disease.

12. The method of claim 1 , wherein the method is used in predictive toxicology for factors including infectious agents that cause or exacerbate Alzheimer’s disease.

13. The method of claim 1 , wherein the method is used to identify causes or accelerators of Alzheimer’s disease.

14. The method of claim 1 , wherein the method is used to identify nutritional factors or supplements for treating Alzheimer’s disease.

15. The method of claim 14, wherein the nutritional factor or supplement is for thiamine or homeostasis of glucose metabolism or other nutritional factors related to pathways regulated by genes identified in Tables 1 , 2, 5 or 7.

16. A patient-specific pharmacotherapeutic method for reducing risk for developing Alzheimer’s disease-associated co-morbidities in a human, the method comprising: a) procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; d) collecting a biological sample from the patient specific neural organoid; e) detecting biomarkers of an Alzheimer's disease related co-morbidity in the patient specific neural organoid sample;

0 administering an anti-Alzheimer’s disease therapeutic agent to the human.

17. The patient specific pharmacotherapeutic method of claim 16, wherein the measured biomarkers comprise biomarkers identified in Table 1, Table 2, Table 5 or Table 7.

18. The method of claim 16 further wherein the measured biomarker is a gene encoding nucleic acids, protein, or their metabolite encoding the biomarkers identified in Table 1, Table 2, Table 5 or Table 7.

19. A plurality of biomarkers comprising a diagnostic panel for predicting a risk for developing Alzheimer’s disease in a human, comprising one or a plurality subset of the biomarkers as identified in Table 1, Table 2, Table 5, or Table 7.

20. The diagnostic panel of claim 19, further wherein the subset of measured biomarkers comprise a gene encoding a nucleic acid, proteins, or their metabolites as identified in Table 1, Table 2, Table 5 or Table 7.

21. A method of pharmaceutical testing for drug screening, toxicity, safety, and/or pharmaceutical efficacy studies using a patient specific neural organoid.

22. A method for detecting at least one biomarker of any of claims 6, 7, 17, 18, 19, or 20, the method comprising: a) obtaining a biological sample from a human patient; and b) contacting the biological sample with an array comprising specific-binding molecules for the at least one biomarker and detecting binding between the at least one biomarker and the specific binding molecules.

23. The method of claim 22, wherein the biomaker is a gene therapy target.

24. A kit comprising an array containing the sequences of one or a plurality of biomarkers of claims 6, 7, 12, 13, 14, or 15 in a human patient.

25. The kit of claim 24 containing a container for collection of a tissue sample from a human. 26. The kit of claim 25 wherein reagents required for RNA isolation from a human tissue sample are included.

27. The kit of claim 24 containing biomarkers for an Alzheimer’s disease genetic disorder.

28. A kit, comprising the container of any of the claims 23-26 and a label or instructions for collection of a sample from a human, isolation of cells, inducement of cells to become pluripotent stem cells, growth of patient-specific neural organoids, isolation of RNA, execution of the array and calculation of gene expression change and prediction of concurrent or future disease risk.

29. The method of claim 1 , wherein the biomarkers are genese encoding nucleic acids, proteins, or their metabolites.

30. A method for detecting one or a plurality of biomarkers from different human chromosomes associated with Alzheimer’s disease or Alzheimer’s disease comorbidity susceptibility using data analytics that obviates the need for whole genome sequence analysis of patient genomes.

31. The method of claim 30, wherein the gene expression level changes are used to determine clinically relevant symptoms and treatments, time of disease onset, and disease severity.

32. The method of claim 30, wherein the neural organoids are used to identify novel biomarkers that serve as data input for development of algorithm techniques as predictive analytics.

33. The method of claim 30, wherein algorithmic techniques include artificial intelligence, machine and deep learning as predictive analytics tools for identifying biomarkers for diagnostic, therapeutic target and drug development process for disease.

34. A method for predicting a risk for developing Alzheimer’s disease in a human, the method comprising: a) procuring one or a plurality of cell samples from the human, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain a neural organoid; d) collecting a biological sample from the neural organoid; e) measuring biomarkers in the neural organoid sample; and

1) detecting measured biomarkers from the neural organoid sample that are differentially expressed in humans with Alzheimer’s disease.

35. The method of claim 34, wherein the at least one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast.

36. The method of claim 34, wherein the measured biomarkers comprise nucleic acids, proteins, or their metabolites.

37. The method of claim 34, wherein the measured biomarker is a nucleic acid encoding human A2M, APR variants.

38. The method of claim 34, wherein the measured biomarkers comprise one or a plurality of genes as identified in Tables 1 , 2, 5 or 6.

39. The method of claim 34, wherein the neural organoid sample is procured from minutes to hours up to 15 weeks post inducement.

40. The method of claim 1 , wherein the biomarkers to be tested are one or a plurality of biomarkers in Table 5.

41. The method of claim 34, wherein the biomarkers to be tested are one or a plurality of biomarkers in Table 6.

42. A method of using a neural organoid along with confirmatory data, and novel data to develop signature algorithms with machine learning, artificial intelligence and deep learning.

43. The method of claim 34, wherein the method is used for diagnostic, therapeutic target discovery and drug action discovery for Alzheimer's disease and Alzheimer’s disease related comorbidites as listed in Table 7. 44. The method of claims 1 , 16, or 34, wherein the neural inventive novel organoid data is corroborated in post mortem tissues from idiopathic patients and extensively identifies known biomarkers for Alzheimer’s disease and comorbidities.

45. The method of claim 44, wherein the method can be used with induced pluripotent stem cells from any skin cell, tissue, or organ from the human body allowing for an all encompassing utility for diagnostics, therapeutic target discovery, and drug development.

46. The method of claims 1 , 16, or 34, wherein the method and/or neural organoid has uses in guided and patient specific toxicology guided by genes from patient’s selective vulnerabililty to infectious agents or to accumulate currently EPA approved safe levels of copper.

47. The method of claim 1 , 16, or 34, wherein the method can be used to identify nutritional and toxicological care that can begin even before birth so that the child develops in utero well.

48. The method of claim 1 , wherein the measured biomarkers comprise nucleic acids, proteins, or their metabolites such as cholesterol.

49. A method for treating Alzheimer’s disease in a human, using a patient-specific pharmacotherapy, the method comprising: a) procuring one or a plurality of cell samples from a healthy human, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more therapeutic patient specific healthy neural organoids; and d) collecting exosomes, and exosome nucleic acids, proteins and metabolites from a plurality of the therapeutic, patient specific healthy neural organoid.

50. The method of claim 49, further comprising: a) procuring one or a plurality of cell samples from a human with Alzheimer's Disease, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more Alzheimer's Disease, patient specific, neural organoids; d) collecting exosome nucleic acid and protein from a plurality of the Alzheimer's Disease patient specific neural organoids; e) detecting changes in Alzheimer's disease exosome nucleic acid and proteins that are differentially expressed in humans with Alzheimer’s disease;

1) performing assays on the Alzheimer’s disease exosome nucleic acids and proteins to identify therapeutic agents that alter the differentially expressed Alzheimer's disease exosome nucleic acids and protein; and g) administering a therapeutic agent to the human with Alzheimer’s disease.

51. The neural organoid of claim 49, wherein the exosome is harvested up to 15 weeks after induction of the neural organoid.

52. The neural organoid of claim 49 wherein the exosome is harvested at minutes, hours, days, or weeks after induction of the neural organoid.

53. The neural organoid of claim 51 , wherein the exosome is harvested at about 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, or 60 minutes after induction of the neural organoid.

54. The neural organoid of claim 51, wherein the exosome is harvested at about 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, or 24 hours after induction of the neural organoid.

55. The neural organoid of claim 51 , wherein the exosome is harvested at about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, or 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks 10 weeks, 11 weeks, 12 weeks or more after induction of the neural organoid.

56. The neural organoid of claim 49, wherein isolated exome nucleic acids and/or proteins are utilized to construct a biomarker library. 57. The neural organoid of claim 56, wherein the isolated exome RNA is used to evaluate the onset or presence of Alzheimer's Disease

58. A method for predicting a risk for developing Alzheimer's disease in a human, the method comprising: a) procuring one or a plurality of cell samples from a healthy human, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more therapeutic patient specific healthy neural organoids; and d) collecting exosome nucleic acids and proteins from a plurality of the therapeutic, patient specific healthy neural organoid.

59. The method of claim 58, further comprising: a) procuring one or a plurality of cell samples from a human with Alzheimer's Disease, comprising one or a plurality of cell types; b) reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; c) treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more Alzheimer's Disease, patient specific, neural organoids; d) collecting exosome nucleic acid and protein from a plurality of the Alzheimer's Disease patient specific neural organoids; e) detecting changes in Alzheimer’s disease exosome nucleic acid and proteins that are differentially expressed in humans with Alzheimer’s disease;

0 performing assays on the Alzheimer’s disease exosome nucleic acids and proteins to identify therapeutic agents that alter the differentially expressed Alzheimer's disease exosome nucleic acids and protein; and g) administering a therapeutic agent to the human with Alzheimer’s disease.

60. The method of claim 58, wherein the measured biomarkers comprise exosome nucleic acids, proteins, or their metabolites .

61. The method of claim 58, wherein the measured biomarker is a nucleic acid encoding human A2M, APP variants. 62. The method of claim 58, wherein the measured biomarkers comprise one or a plurality of genes as identified in Tables 1 , 2, 5 or 6.

63. The method of claim 58, wherein the neural organoid sample is procured from minutes to hours up to 15 weeks post inducement.

64. The method of claim 58, wherein the biomarkers to be tested are one or a plurality of biomarkers in Table 5.

65. The method of claim 58, wherein the biomarkers to be tested are one or a plurality of biomarkers in Table 6.

Description:
NEURAL DERIVED EXOSOMES FOR ALZHEIMER’S DISEASE AND COMORBIDITIES THEREOF

FIELD OF THE INVENTION

[0001] This disclosure relates to production and use of human stem cell derived neural organoids to identify patients with Alzheimer’s disease and Alzheimer's disease patient treatment using patient-specific pharmacotherapy. Further disclosed are patient-specific pharmacotherapeutic methods for reducing risk for developing Alzheimer’s disease- associated co-morbidities in a human. Also disclosed are methods to predict onset risk of Alzheimer's disease (and identified comorbidities) in an individual. In particular, the inventive processes disclosed herein provide neural organoid reagents produced from an individual's induced pluripotent stem cells (iPSCs) for identifying patient-specific pharmacotherapy, predictive biomarkers, and developmental and pathogenic gene expression patterns and dysregulation thereof in disease onset and progression, and methods for diagnosing prospective and concurrent risk of development or establishment of Alzheimer’s disease (and comorbidities) in the individual. The invention also provides reagents and methods for identifying, testing, and validating therapeutic modalities, including chemical and biologic molecules for use as drugs for ameliorating or curing Alzheimer’s disease.

BACKGROUND OF THE INVENTION

[0002] The human brain, and diseases associated with it have been the object of investigation and study by scientists for decades. Throughout this time, neurobiologists have attempted to increase their understanding of the brain's capabilities and functions. Neuroscience has typically relied on the experimental manipulation of living brains or tissue samples, but a number of factors have limited scientific progress. For ethical and practical reasons, obtaining human brain tissue is difficult while most invasive techniques are impossible to use on live humans. Experiments in animals are expensive and time- consuming and many animal experiments are conducted in rodents, which have a brain structure and development that vary greatly from humans. Results obtained in animals must be verified in long and expensive human clinical trials and much of the time the animal disease models are not folly representative of disease pathology in the human brain.

[0003] Improved experimental models of the human brain are urgently required to understand disease mechanisms and test potential therapeutics. The ability to detect and diagnose various neurological diseases in their early stages could prove critical in the effective management of such diseases, both at times before disease symptoms appear and thereafter. Neuropathology is a frequently used diagnostic method; however, neuropathology is usually based on autopsy results. Molecular diagnostics promises to provide a basis for early detection and a risk of early onset of neurological disease.

However, molecular diagnostic methods in neurological diseases are limited in accuracy, specificity, and sensitivity. Therefore, there is a need in the art for non-invasive, patient specific molecular diagnostic methods to be developed.

[0004] Consistent with this need, neural organoids hold significant promise for studying neurological diseases and disorders. Neural organoids are developed from cell lineages that have been first been induced to become pluripotent stem cells. Thus, the neural organoid is patient specific. Importantly, such models provide a method for studying neurological diseases and disorders that overcome previous limitations. Accordingly, there is a need in the art to develop patient-specific reagents, therapeutic modalities, and methods based on predictive biomarkers for diagnosing and/or treating current and future risk of neurological diseases including Alzheimer’s disease.

SUMMARY OF THE INVENTION

[0005] This disclosure, in one embodiment, provides neural reagents and methods for treating Alzheimer’s disease in a human, using patient-specific pharmacotherapies, the methods comprising: procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; detecting changes in Alzheimer’s disease biomarker expression from the patient specific neural organoid sample that are differentially expressed in humans with Alzheimer’s disease; performing assays on the patient specific neural organoid to identify therapeutic agents that alter the differentially expressed Alzheimer’s disease biomarkers in the patient-specific neural organoid sample; and administering a therapeutic agent for Alzheimer’s disease to treat the human. In one aspect at least one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast derived from skin or blood cells from humans. In another aspect the fibroblast derived skin or blood cells from humans is identified with the genes identified in Table 1 (Novel Alzheimer’s disease Biomarkers), Table 2 (Biomarkers for Alzheimer’s disease), Table 5 (Alzheimer’s disease Therapeutic Neural Organoid Authentication Genes), or Table 7 (Genes and Accession Numbers for Co-Morbidity Susceptibility / Resistance Associated with Alzheimer’s disease). In yet another aspect, the measured biomarkers comprise nucleic acids, proteins, or their metabolites. In another aspect the measured biomarkers comprise one or a plurality of biomarkers identified in Table 1 , Table 2, Table 5 or Table 7 or variants thereof. In yet another aspect, a combination of biomarkers is detected, the combination comprising a nucleic acid encoding human A2M, APR variants and one or a plurality of biomarkers comprising a nucleic acid encoding human genes identified in Table 1.

[0006] In one aspect of the disclosure, the biomarkers for Alzheimer’s disease include human nucleic acids, proteins, or their metabolites as listed in Table 1.

TABLE 1. Novel Alzheimer's disease Biomarkers

[0007] One of skill in the art will recognize that sequence data for the genes listed above can be obtained in publicly available gene databases such as GeneCards, GenBank, Malcard, Uniport and PathCard databases.

[0008] In still another aspect, the neural organoid biological sample is collected after about one hour up to about 12 weeks post inducement. In another aspect the neural organoid sample is procured from structures of the neural organoid that mimic structures developed in utero at about 5 weeks. In yet another aspect the neural organoid at about twelve weeks post-inducement comprises structures and cell types of retina, cortex, midbrain, hindbrain, brain stem, or spinal cord. In a one aspect the neural organoid contains microglia, and one or a plurality of Alzheimer’s disease biomarkers as identified in Table 1 and Table 7. [0009] In yet another aspect the method is used to detect environmental factor susceptibility including infectious agents that cause or exacerbate Alzheimer's disease, or accelerators of Alzheimer's disease. In a further aspect the method is used to identify nutritional factor deficiency susceptibility or supplements for treating Alzheimer’s disease. In a further aspect the nutritional factor or supplement is for glucose dyshometostasis or other nutritional factors related to pathways (Pathcards database; Weizmann Institute of Science) regulated by genes identified in Tables 1, 2, 5 or 7. In yet another aspect fetal cells from amniotic fluid can be used to grow neural organoids and as such nutritional and toxicological care can begin even before birth so that the child develops in utero well.

[0010] In a second embodiment, the disclosure provides methods for reducing risk of developing Alzheimer’s disease associated co-morbidities in a human comprising procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; detecting biomarkers of an Alzheimer’s disease related co-morbidity in the patient specific neural organoid sample that are differentially expressed in humans with Alzheimer’s disease; and administering an anti- Alzheimer’s or anti co-morbidity therapeutic agent to the human.

In one aspect the measured biomarkers comprise biomarkers identified in Table 1, Table 2, Table 5 or Table 7 and can be nucleic acids, proteins, or their metabolites (identifiable in GeneCards and PathCard databases). In a further aspect the invention provides diagnostic methods for predicting risk for developing Alzheimer’s disease in a human, comprising one or a plurality subset of the biomarkers as identified in Table 1, Table 2, Table 5, or Table 7.

In yet another aspect, the subset of measured biomarkers comprise nucleic acids, proteins, or their metabolites as identified in Table 1 , Table 2, Table 5 or Table 7. The biomarkers can be co related to disease onset, progression, and severity and include glucose, and cholesterol mentabolism.. In another aspect the method and/or neural organoid has uses in guided and patient specific toxicology guided by genes from patient’s selective vulnerabililty to infectious agents or to accumulate currently EPA approved safe levels of copper.

[0011] In another embodiment are methods of pharmaceutical testing for Alzheimer’s disease drug screening, toxicity, safety, and/or pharmaceutical efficacy studies using patient- specific neural organoids.

[0012] In an additional embodiment, methods are provided for detecting at least one biomarker of Alzheimer’s disease, the method comprising, obtaining a biological sample from a human patient; and contacting the biological sample with an array comprising specific-binding molecules for the at least one biomarker and detecting binding between the at least one biomarker and the specific binding molecules.

[0013] In one aspect the biomaker detected is a gene therapy target.

[0014] In a further embodiment the disclosure provides a kit comprising an array containing sequences of biomarkers from Table 1 or Table 2 for use in a human patient. In one aspect, the kit further contains reagents for RNA isolation and biomarkers for Alzheimer’s disease.

In a further aspect, the kit further advantageously comprises a container and a label or instructions for collection of a sample from a human, isolation of cells, inducement of cells to become pluripotent stem cells, growth of patient-specific neural organoids, isolation of RNA, execution of the array and calculation of gene expression change and prediction of concurrent or future disease risk. In one aspect of the disclosure, the biomarkers for Alzheimer’s disease include human nucleic acids, proteins, or their metabolites as listed in Table 1.

[0015] In one aspect, the biomarkers can include biomarkers listed in Table 2. In another aspect, biomarkers can comprise any markers or combination of markers in Tables 1 and 2 or variants thereof.

TABLE 2. Biomarkers for Alzheimer’s Disease

[0016] One of skill in the art will recognize that sequence data for the genes listed above can be obtained in publicly available gene databases such as GeneCards, GenBank, Malcard, Uniport and PathCard databases.

[0017] In a further embodiment, the disclosure provides a method for detecting one or a plurality of biomarkers from different human chromosomes associated with Alzheimer's disease or Alzheimer's disease comorbidity susceptibility using data analytics that obviates the need for whole genome sequence analysis of a person or patient’s genome. In one aspect the methods are used to determine gene expression level changes that are used to identifly clinically relevant symptoms and treatments, time of disease onset, and disease severity. In yet another aspect the neural organoids are used to identify novel biomarkers that serve as data input for development of algorithm techniques as predictive analytics. In a further aspect the algorithmic techniques include artificial intelligence, machine and deep learning as predictive analytics tools for identifying biomarkers for diagnostic, therapeutic target and drug development process for disease. In one aspect the neural neural organoid along with confirmatory data, and novel data can be used to develop signature algorithms with machine learning, artifiical intelligence and deep learning. In another aspect the the method is used for diagnostic, therapeutic target discovery and drug action discovery for Alzheimer’s disease and Alzheimer’s disease related comorbidites as listed in Table 7. In yet another aspect the inventive model neural organoid data is corroborated in post mortem tissues from idiopathic patients and extensively identifies known biomarkers for Alzheimer’s disease and comorbidities. In yet another aspect the method can be used with induced pluripotent stem cells from any skin cell, tissue, or organ from the human body allowing for an all encompassing utility for diagnostics, therapeutic target discovery, and drug development. [0018] In yet another embodiment the invention provides methods for predicting a risk comorbidity onset that accompanies Alzheimer’s disease. Said methods first determines gene expression changes in neural organoids from a normal human individual versus a human individual with Alzheimer's disease. Genes that change greater than 1.4 fold are associated with co-morbidities as understood by those skilled in the art.

[0019] In a further embodiment, the invention provides kits for predicting the risk of current or future onset of Alzheimer’s disease. Said kits provide reagents and methods for identifying from a patient sample gene expression changes for one or a plurality of disease- informative genes for individuals without a neurological disease that is Alzheimer’s disease. [0020] In an additional embodiment, the invention provides methods for identifying therapeutic agents for treating Alzheimer’s disease. Such embodiments comprise using the neural organoids provided herein, particularly, but not limited to said neural organoids from iPSCs from an individual or from a plurality or population of individuals. The inventive methods include assays on said neural organoids to identify therapeutic agents that alter disease-associated changes in gene expression of genes identified as having altered expression patterns in disease, so as to express gene expression patterns more closely resembling expression patterns for disease-informative genes for individuals without a neurological disease that is Alzheimer's disease.

[0021] In another embodiment, the invention provides methods for predicting a risk for developing Alzheimer’s disease in a human, comprising procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; measuring biomarkers in the neural organoid sample; and detecting measured biomarkers from the neural organoid sample that are differentially expressed in humans with Alzheimer’s disease. In certain embodiments, the at least one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast. In certain embodiments, the measured biomarkers comprise nucleic acids, proteins, or their metabolites. In certain embodiments, the measured biomarker is a nucleic acid encoding human A2M and APP variants. In certain embodiments, the measured biomarkers comprise one ora plurality of genes as identified in Tables 1, 2, 5 or 6. In certain embodiments, the neural organoid sample is procured from minutes to hours up to 15 weeks post inducement. In certain embodiments, the biomarkers to be tested are one or a plurality of biomarkers in Tables 5 or 6 (Alzheimer’s disease Diagnostic Neural Organoid Authentication Genes). [0022] These and other data findings, features, and advantages of the present invention will be more fully understood from the following detailed description taken together with the accompanying claims. It is noted that the scope of the claims is defined by the recitations therein and not by the specific discussion of features and advantages set forth in the present description.

BRIEF DESCRIPTION OF THE FIGURES

[0023] Fig 1A is a micrograph showing a 4X dark field image of Brain Organoid Structures typical of approximately 5-week in utero development achieved in 12 weeks in vitro. Average size: 2-3 mm long. A brain atlas is provided for reference (left side).

[0024] FIG. 1 B shows immuno-fluorescence images of sections of iPSC-derived human brain organoid after approximately 12 weeks in culture. Z-stack of thirty-three optical sections, 0.3 microns thick were obtained using laser confocal imaging with a 40X lens. Stained with Top panel: beta III tubulin (green: axons); MAP2 (red: dendrites); Hoechst (blue: nuclei); Bottom panel: Doublecortin (red).

[0025] FIG. 2 is a micrograph showing immunohistochemical staining of brain organoid section with the midbrain marker tyrosine hydroxylase. Paraformaldehyde fixed sections of a 8- week old brain organoid was stained with an antibody to tyrosine hydroxylase and detected with Alexa 488 conjugated secondary Abs (green) and counter stained with Hoechst to mark cell nuclei (blue). Spinning disc confocal image (40X lens) of section stained with an antibody that binds tyrosine hydroxylase and Hoechst (scale bar 10pm). [0026] FIG. 3: Spinning disc confocal image (40X lens) of section. Astrocytes stained with GFAP (red) and mature neurons with NeuN (green).

[0027] FIG. 4 is a schematic showing in the upper panel a Developmental Expression Profile for transcripts as Heat Maps ofNKCC 1 and KCC2 expression at week 1, 4 and 12 of organoid culture as compared to approximate known profiles (lower panel). NKCCI: Na(+)- K(+)-CI(-) cotransporter isoform 1. KCC2: K(+)-CI(-) cotransporter isoform 2.

[0028] Fig 5A is a schematic showing GABAergic chloride gradient regulation by NKCC 1 and KCC2.

[0029] FIG. 5B provides a table showing a representative part of the entire transcriptomic profile of brain organoids in culture for 12 weeks measured using a transcriptome sequencing approach that is commercially available (AmpliSeq™). The table highlights the expression of neuronal markers for diverse populations of neurons and other cell types that are comparable to those expressed in an adult human brain reference (HBR; Clontech) and the publicly available embryonic human brain (BRAINS CAN) atlas of the Allen Institute database.

[0030] FIG. 5C provides a table showing AmpliSeq Tm gene expression data comparing gene expression in an organoid (column 2) at 12 weeks in vitro versus Human Brain Reference (HBR; column 3). A concordance of greater than 98% was observed. [0031] FIG. 5D provides a table showing AmpliSeq™ gene expression data comparing organoids generated during two independent experiments after 12 weeks in culture (column

2 and 3). Gene expression reproducibility between the two organoids was greater than 99%. Note that values are CPM (Counts Per Kilo Base per Million reads) in the tables and <1 is background.

[0032] FIG. 6A is a schematic showing results of developmental transcriptomics. Brain organoid development in vitro follows KNOWN Boolean logic for the expression pattern of transcription factors during initiation of developmental programs of the brain. Time Points: 1, 4, and 12 Weeks. PITX3 and NURRI (NR4A) are transcription factors that initiate midbrain development (early; at week 1), DLKI, KLHLI, PTPRU, and ADH2 respond to these two transcription factors to further promote midbrain development (mid; at week 4 &12), and TH, VMAT2, DAT and D2R define dopamine neuron functions mimicking in vivo development expression patterns. The organoid expresses genes previously known to be involved in the development of dopaminergic neurons (Blaess S, Ang SL. Genetic control of midbrain dopaminergic neuron development. Wiley Interdiscip Rev Dev Biol. 2015 Jan 6. doi: 10.1002/wdev. I69).

[0033] FIGs. 6B-6D are tables showing AmpliSeq™ gene expression data for genes not expressed in organoid (column 2 in 6B, 6C, and 6D) and Human Brain Reference (column

3 in 6B, 6C, and 6D). This data indicates that the organoids generated do not express genes that are characteristic of non-neural tissues. This gene expression concordance is less than 5% for approximately 800 genes that are considered highly enriched or specifically expressed in a non-neural tissue. The olfactory receptor genes expressed in the olfactory epithelium shown are a representative example. Gene expression for most genes in table is less than one or zero.

[0034] FIG. 7 includes schematics showing developmental heat maps of transcription factors (TF) expressed in cerebellum development and of specific Markers GRID 2.

[0035] FIG. 8 provides a schematic and a developmental heat map of transcription factors expressed in Hippocampus Dentate Gyms.

[0036] FIG. 9 provides a schematic and a developmental heat map of transcription factors expressed in GABAergic Intemeuron Development. GABAergic Intemeurons develop late in vitro.

[0037] FIG. 10 provides a schematic and a developmental heat map of transcription factors expressed in Serotonergic Raphe Nucleus Markers of the Pons.

[0038] FIG. 11 provides a schematic and a developmental heat map of transcription factor transcriptomics (FIG. 11 A). Hox genes involved in spinal cord cervical, thoracic, and lumbar region segmentation are expressed at discrete times in utero. The expression pattern of these Hoxgene in organoids as a function of in vitro developmental time (1 week; 4 weeks; 12 weeks; ; Figures 11 B and 11C)

[0039] FIG. 12 is a graph showing the replicability of brain organoid development from two independent experiments. Transcriptomic results were obtained by Ampliseq analysis of normal 12-weekoti brain organoids. The coefficient of determination was 0.6539.

[0040] FIG. 13 provides a schematic and gene expression quantification of markers for astrocytes, oligodendrocytes, microglia, and vasculature cells.

[0041] FIG. 14 shows developmental heat maps of transcription factors (TF) expressed in retina development and other specific Markers. Retinal markers are described, for example, in Farkas et al. BMC Genomics 2013, 14:486.

[0042] FIG. 15 shows developmental heat maps of transcription factors (TF) and Markers expressed in radial glial cells and neurons of the cortex during development [0043] FIG. 16 is a schematic showing the brain organoid development in vitro. iPSC stands for induced pluripotent stem cells. NPC stands for neural progenitor cell.

[0044] FIG. 17 is a graph showing the replicability of brain organoid development from two independent experiments.

[0045] FIGS. 18A and 18B are tables showing the change in the expression level of certain genes in APP gene duplication organoid.

[0046] FIG. 19 is human genetic and postmortem brain analysis published data that independently corroborate biomarkers predicted from the Alzheimer’s disease neural organoid derived data, including novel changes in microglial functions increasing susceptibility to infectious agents in Alzheimer's disease.

DETAILED DESCRIPTION

[0047] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. The following references provide one of skill with a general definition of many of the terms used in this disclosure: Singleton etal., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Vtalker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Veriag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). These references are intended to be exemplary and illustrative and not limiting as to the source of information known to the worker of ordinary skill in this art. As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise.

[0048] It is noted here that as used in this specification and the appended claims, the singular forms "a," "an," and "the” also include plural reference, unless the context clarity dictates otherwise. [0049] The term ‘about" or “approximately" means within 25%, such as within 20% (or 5% or less) of a given value or range.

[0050] As used herein, the terms “or” and “and/or * are utilized to describe multiple components in combination or exclusive of one another. For example, “x, y, and/or z" can refer to “x" alone, “y * alone, “z” alone, “x, y, and z,” “(x and y) or z,” “x or (y and z),“ or “x or y or z." [0051] It is noted that terms like “preferably,” “commonly,” and “typically” are not utilized herein to limit the scope of the claimed invention or to imply that certain features are critical, essential, or even important to the structure or function of the claimed invention. Rather, these terms are merely intended to highlight alternative or additional features that can or cannot be utilized in a particular embodiment of the present invention.

[0052] For the purposes of describing and defining the present invention, it is noted that the term “substantially" is utilized herein to represent the inherent degree of uncertainty that can be attributed to any quantitative comparison, value, measurement, or other representation. The term “substantially" is also utilized herein to represent the degree by which a quantitative representation can vary from a stated reference without resulting in a change in the basic function of the subject matter at issue.

[0053] A "neural organoid” means a non-naturally occurring three-dimensional organized cell mass that is cultured in vitro from a human induced pluripotent stem cell and develops similaly to the human nervous system in terms of neural marker expression and structure. Further a neural organoid has two or more regions. The first region expresses cortical or retinal marker or markers. The remaining regions each express markers of the brain stem, cerebellum, and/or spinal cord.

[0054] Neural markers are any protein or polynucleotide expressed consistent with a cell lineage. By "heural marker" it is meant any protein or polynucleotide, the expression of which is associated with a neural cell fate. Exemplary neural markers include markers associated with the hindbrain, midbrain, forebrain, or spinal cord. One skilled in the art will understand that neural markers are representative of the cerebrum, cerebellum and brainstem regions. Exemplary brain structures that express neural markers include the cortex, hyopthalamus, thalamus, retina, medulla, pons, and lateral ventricles. Further, one skilled in the art will recognize that within the brain regions and structures, granular neurons, dopaminergic neurons, GABAergic neurons, cholinergic neurons, glutamatergic neurons, serotonergic neurons, dendrites, axons, neurons, neuronal, cilia, purkinje fibers, pyramidal cells, spindle cells, express neuronal markers. One skilled in the art will recognize that this list is not all encompassing and that neural markers are found throughout the central nervous system including other brain regions, structures, and cell types.

[0055] Exemplary cerebellar markers include but are not limited to ATOH1 , PAX6, SOX2, LHX2, and GRID2. Exemplary markers of dopaminergic neurons include but are not limited to tyrosine hydroxylase, vesicular monoamine transporter 2 (VMAT2), dopamine active transporter (DAT) and Dopamine receptor D2 (D2R). Exemplary cortical markers include, but are not limited to, doublecortin, NeuN, FOXP2, CNTN4, and TBR1. Exemplary retinal markers include but are not limited to retina specific Guanylate Cyclases (GUY2D, GUY2F), Retina and Anterior Neural Fold Homeobox (RAX), and retina specific Amine Oxidase, Copper Containing 2 (RAX). Exemplary granular neuron markers include, but are not limited to SOX2, NeuroDI , DCX, EMX2, FOXG1I, and PROX1. Exemplary brain stem markers include, but are not limited to FGF8, INSM1 , GATA2, ASCLI, GATA3. Exemplary spinal cord markers include, but are not limited to homeobox genes including but not limited to HOXA1 ,

HOXA2, HOXA3, HOXB4, HOXA5, HOXCS, or HOXDI3. Exemplary GABAergic markers include, but are not limited to NKCCI or KCC2. Exemplary astrocytic markers include, but are not limited to GFAP. Exemplary oliogodendrocytic markers include, but are not limited to OLIG2 or MBP. Exemplary microglia markers include, but are not limited to AIF1 or CD4. In one embodiment the measured biomarkers listed above have at least 70% homology to the sequences in the Appendix. One skilled in the art will understand that the list is exemplary and that additional biomarkers exist.

[0056] Diagnostic or informative alteration or change in a biomarker is meant as an increase or decrease in expression level or activity of a gene or gene product as detected by conventional methods known in the art such as those described herein. As used herein, such an alteration can include a 10% change in expression levels, a 25% change, a 40% change, or even a 50% or greater change in expression levels.

[0057] A mutation is meant to include a change in one or more nucleotides in a nucleotide sequence, particularly one that changes an amino acid residue in the gene product. The change may or may not have an impact (negative or positive) on activity of the gene.

NEURAL ORGANOIDS

[0058] Neural organoids are generated in vitro from patient tissue samples. Neural organoids were previously disclosed in WO2017123791 A1 (https://patents.google.com/patent/WO2017123791A1/en), incorporated herein, in its entirety. A variety of tissues can be used including skin cells, hematopoietic cells, or peripheral blood mononuclear cells (PBMCs) or in vivo stem cells directly. One of skill in the art will further recognize that other tissue samples can be used to generate neural organoids. Use of neural organoids permits study of neural development in vitro. In one embodiment skin cells are collected in a petri dish and induced to an embryonic-like pluripotent stem cell (iPSC) that have high levels of developmental plasticity. iPSCs are grown into neural organoids in said culture under appropriate conditions as set forth herein and the resulting neural organoids closely resemble developmental patterns similar to human brain. In particular, neural organoids develop anatomical features of the retina, forebrain, midbrain, hindbrain, and spinal cord. Importantly, neural organoids express >98% of the about 15,000 transcripts found in the adult human brain. iPSCs can be derived from the skin or blood cells of humans identified with the genes listed in Table 1 (Novel Markers of Alzheimer's disease), Table 2 (Markers of Alzheimer’s disease), Table 5 (Neural Organoid Alzheimer’s disease Authenticating Genes) and Table 7 (Comorbidities of Alzheimer’s disease).

[0059] In one embodiment, the about 12-week old iPSC-derived human neural organoid has ventricles and other anatomical features characteristic of a 35-40 day old neonate. In an additional embodiment the about 12 week old neural organoid expresses beta 3-tubulin, a marker of axons as well as somato-dendritic Puncta staining for MAP2, consistent with dendrites. In yet another embodiment, at about 12 weeks the neural organoid displays laminar organization of cortical structures. Cells within the laminar structure stain positive for doublecortin (cortical neuron cytosol), Beta3 tubulin (axons) and nuclear staining. The neural organoid, by 12 weeks, also displays dopaminergic neurons and astrocytes.

[0060] Accordingly as noted, neural organoids permit study of human neural development in vitro. Further, the neural organoid offers the advantages of replicability, reliability and robustness, as shown herein using replicate neural organoids from the same source of iPSCs.

DEVELOPMENTAL TRANSCRIPTOMICS

[0061] A “transcriptome” is a collection of all RNA, including messenger RNA (mRNA), long non-coding RNAs (IncRNA), microRNAs (miRNA) and, small nucleolar RNA snoRNA), other regulatory polynucleotides, and regulatory RNA (IncRNA, miRNA) molecules expressed from the genome of an organism through transcription therefrom. Thus, transcriptomics is the study of the mRNA transcripts produced by the genome at a given time in any particular cell or tissue of the organism. Transcriptomics employs high-throughput techniques to analyze genome expression changes associated with development or disease. In certain embodiments, transcriptomic studies can be used to compare normal, healthy tissues and diseased tissue gene expression. In further embodiments, mutated genes or variants associated with disease or the environment can be identified.

[0062] Consistent with this, the aim of developmental transcriptomics is identifying genes associated with, or significant in, organismal development and disease and dysfunctions associated with development. During development, genes undergo up- and down-regulation as the organism develops. Thus, transcriptomics provides insight into cellular processes, and the biology of the organism. [0063] Generally, in one embodiment RNA is sampled from the neural organoid described herein within at about one week, about four weeks, or about twelve weeks of development; most particularly RNA from all three time periods are samples. However, RNA from the neural organoid can be harvested at minutes, hours, days, or weeks after reprogramming. For instance, RNA can be harvested at about 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, and 60 minutes. In a further embodiment the RNA can be harvested 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, or 24 hours. In a further embodiment the RNA can be harvested at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, or 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks 10 weeks, 11 weeks, 12 weeks or more in culture. After enriching for RNA sequences, an expressed sequence tag (EST) library is generated and quantitated using the AmpliSeq™ technique from ThermoFisher. Exemplars of alternate technologies include RNASeq and chip based hybridization methods. Transcript abundance in such experiments is compared in control neural organoids from healthy individuals vs. neural organoids generated from individuals with disease and the fold change in gene expression calculated and reported.

[0064] Furthermore, in one embodiment RNA from neural organoids for Alzheimer’s disease, are converted to DNA libraries and then the representative DNA libraries are sequenced using exon-specific primers for 20,814 genes using the AmpliSeq™ technique available commercially from ThermoFisher. Reads in cpm <1 are considered background noise. All cpm data are normalized data and the reads are a direct representation of the abundance of the RNA for each gene.

[0065] Briefly, in one embodiment, the array consists of one or a plurality of genes used to predict risk of Alzheimer’s disease. In an alternative embodiment, reads contain a plurality of genes that are used to treat Alzheimer’s disease in a human, using patient-specific pharmacotherapy known to be associated with Alzheimer's disease. In one aspect, the gene libraries can be comprised of disease-specific gene as provided in Tables 1 and 2 or a combination of genes in Table 1 or Table 2 with alternative disease specific genes. Exemplarily, changes in expression or mutation of disease-specific genes are detected using such sequencing, and differential gene expression detected thereby, qualitatively by detecting a pattern of gene expression or quantitatively by detecting the amount or extent of expression of one or a plurality of disease-specific genes or mutations thereof. Results of said assays using the AmpliSeq™ technique can be used to identify genes that can predict disease risk or onset and can be targets of therapeutic intervention. In further embodiments, hybridization assays can be used, including but not limited to sandwich hybridization assays, competitive hybridization assays, hybridization-ligation assays, dual ligation hybridization assays, or nuclease assays.

Neural Organoids and Pharmaceutical Testing

[0066] Neural organoids are useful for pharmaceutical testing. Currently, drug screening studies including toxicity, safety and or pharmaceutical efficacy, are performed using a combination of in vitro work, rodent / primate studies and computer modeling. Collectively, these studies seek to model human responses, in particular physiological responses of the central nervous system.

[0067] Human neural organoids are advantageous over current pharmaceutical testing methods for several reasons. First neural organoids are easily derived from healthy and diseased patients, mitigating the need to conduct expensive clinical trials. Second, rodent models of human disease are unable to mimic physiological nuances unique to human growth and development. Third, use of primates creates ethical concerns. Finally, current methods are indirect indices of drug safety. Alternatively, neural organoids offer an inexpensive, easily accessible model of human brain development. This model permits direct, and thus more thorough, understanding of the safety, efficacy, and toxicity of pharmaceutical compounds.

[0068] Starting material for neural organoids is easily obtained from healthy and diseased patients. Further, because human organoids are easily grown they can be produced en mass. This permits efficient screening of pharmaceutical compounds.

[0069] Neural organoids are advantageous for identifying biomarkers of a disease or a condition, the method comprising a) obtaining a biological sample from a human patient; and b) detecting whether at least one biomarker is present in the biological sample by contacting the biological sample with an array comprising binding molecules specific for the biomarkers and detecting binding between the at least one biomarker and the specific binding molecules. In further embodiments, the biomarker serves as a gene therapy target.

DEVELOPMENTAL TRANSCRIPTOMICS AND PREDICTIVE MEDICINE [0070] Changes in gene expression of specific genes when compared to those from non- diseased samples by >1.4 fold identify candidate genes correlating with a disease. Further searches of these genes in data base searches (e.g. Genecard, Malacard, Pubmed; Human Protein Atlas (https://www.proteinatlas.org/ENSG00000115091-ACTR3/patholog y) identify known diseases correlated previously with the disease state. In one embodiment AmpliSeq™ quantification of fold expression change allows for determination of fold change from control. ALZHEIMER'S DISEASE

[0071] Alzheimer's Disease (AD) is an irreversible brain disorder. The disease is a common form of dementia, is associated with memory loss and interferes with other intellectual abilities that complicate daily life. Alzheimer's disease accounts for 60 to 80 percent of dementia cases. Disease onset occurs most often for individuals in their mid-60s and is estimated to affect approximately five million individuals at present. However, disease onset occurs many years prior to physical expression of symptoms. The cost to society currently exceeds $270 billion and no effective treatment currently exists.

[0072] The etiology of AD is thought to involve two abnormal structures, plaques and tangles, that damage and kill nerve cells in human brain. Plaques are deposits of beta- amyloid protein fragments that build up in the spaces between nerve cells, while tangles are twisted fibers of tau, a protein that builds up inside cells. In addition, anatomical examination reveals a loss of neuronal connections in most AD patients. The result is a loss of cognitive function and the ability to perform easily normal daily activities. Thus, AD patients need extensive caregiver assistance. As a result AD is a significant financial, physical and emotional burden and one of the top causes of death in the United States.

[0073] AD diagnosis often occurs after the onset of physical symptoms. Individuals at risk for AD would benefit from earlier detection of the disease. In addition, early detection of AD would permit development of pharmaceutical and related treatments to improve AD-related outcomes and delay disease onset. This disclosure provides, in a first embodiment, neural reagents and methods for treating Alzheimer’s disease in a human, using patient-specific pharmacotherapies, the methods comprising: procuring one or a plurality of cell samples from a human, comprising one ora plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; detecting changes in Alzheimer's disease biomarker expression from the patient specific neural organoid sample that are differentially expressed in humans with Alzheimer’s disease; performing assays on the patient specific neural organoid to identify therapeutic agents that alter the differentially expressed Alzheimer’s disease biomarkers in the patient-specific neural organoid sample; and administering a therapeutic agent for Alzheimer’s disease to treat the human.

[0074] In one aspect at least one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast derived from skin or blood cells from humans. In another aspect the fibroblast derived skin or blood cells from humans is identified with the genes identified in Table 1 (Novel Alzheimer’s disease Biomarkers), Table 2 (Biomarkers for Alzheimer’s disease), Table 5 (Therapeutic Neural Organoid Authentication Genes), or Table 7 (Genes and Acession Numbers for Co-Morbidities Associated with Alzheimer’s disease). In yet another aspect, the measured biomarkers comprise nucleic acids, proteins, or their metabolites. In another aspect the measured biomarkers comprise one or a plurality of biomarkers identified in Table 1 , Table 2, Table 5 or Table 7 or variants thereof. In yet another aspect, a combination of biomarkers is detected, the combination comprising a nucleic acid encoding human A2M, APP variants; and one or a plurality of biomarkers comprising a nucleic acid encoding human genes identified in Table 1.

[0075) In one aspect of the disclosure, the biomarkers for Alzheimer’s disease include human nucleic acids, proteins, or their metabolites as listed in Table 1. These are biomarkers that are found to change along with numerous others ones that are extensively correlated with postmortem brains from Alzheimer’s disease patients.

[0076] In still another aspect, the neural organoid biological sample is collected after about one hour up to about 12 weeks post inducement. In another aspect the neural organoid sample is procured from structures of the neural organoid that mimic structures developed in utero at about 5 weeks. In yet another aspect the neural organoid at about twelve weeks post-inducement comprises structures and cell types of retina, cortex, midbrain, hindbrain, brain stem, or spinal cord. In a one aspect the neural organoid contains microglia, and one or a plurality of Alzheimer’s disease biomarkers as identified in Table 1 and Table 7. In yet another aspect the method is used to detect environmental factors such as infectious agents that cause or exacerbate Alzheimer’s disease, or accelerators of Alzheimer’s disease. An accelerator of Alzheimer’s disease is an environmental or nutritional factor that specifically interacts with an Alzheimer’s disease specific biomarker to affect downstream process related to these biomarkers biological function such that a subclinical or milder state of Alzheimer’s disease becomes a full blown clinical state earlier or more severe in nature. These can be determined, without whole genome sequence analysis of patient genomes, solely from comparative differential gene expression analyses of in vitro neural organoids as models of brain development, only in conjunction with an inventive process that reproducibly and robustly promotes development of all the major brain regions and cell types.

[0077] The detection of novel biomarkers, as presented in Table 1 and/or Tables 2, 5, and 6 can be used to identity individuals who should be provided prophylactic treatment for Alzheimer’s disease. In one aspect such treatments can include avoidance of environmental stimuli and accelerators that exacerbate Alzheimer’s disease. In a further aspect early diagnosis can be used in a personalized medicine approach to identify new patient specific pharmacotherapies for Alzheimer’s disease based on biomarker data. In a further aspect, the neural organoid model can be used to test the effectiveness of currently utilized Alzheimer’s disease therapies. In one aspect the neural organoid can be used to identify the risk and/or onset of Alzheimer’s disease and additionally, provide patient-specific insights into the efficacy of using known pharmacological agents to treat Alzheimer’s disease. This allows medical professionals to identify and determine the most effective treatment for an individual Alzherimer's disease patient, before symptoms arise. Furthermore, one skilled in the art will recognize that the effectiveness of additional FDA-approved, as well as novel drugs underdevelopment could be tested using the methods disclose herein. In a further aspect the method allows for development and testing of non-individualized, global treatment strategies for mitigating the effects and onset of Alzheimer’s disease.

[0078] In a further aspect the method is used to identify nutritional factors or supplements for treating Alzheimer’s disease. In a further aspect the nutritional factor or supplement is thiamine or glucose homeostasis or other nutritional factors related to pathways regulated by genes identified in Tables 1 , 2, 5 or 7.

[0079] In a second embodiment, the disclosure provides methods for reducing risk of developing Alzheimer’s disease associated co-morbidities in a human comprising procuring one or a plurality of cell samples from a human, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; detecting changes in Alzheimer’s disease biomarker expression from the patient specific neural organoid sample that are differentially expressed in humans with Alzheimer’s disease; and administering a therapeutic agent to treat Alzheimer’s disease. In one aspect the measured biomarkers comprise biomarkers identified in Table 1, Table 2, Table 5 or Table 7 and can be genes, proteins, or their metabolites.

[0080] In a third embodiment, the disclosure provides diagnostic methods for predicting risk for developing Alzheimer’s disease in a human, comprising one or a plurality subset of the biomarkers as identified in Table 1 , Table 2, Table 5, or Table 7. In yet another aspect, the subset of measured biomarkers comprise nucleic acids, proteins, or their metabolites as identified in Table 1 , Table 2, Table 5 or Table 7.

[0081] In a fourth embodiment are methods of pharmaceutical testing for Alzheimer’s disease drug screening, toxicity, safety, and/or pharmaceutical efficacy studies using patient-specific neural organoids.

[0082] In a fifth embodiment, methods are provided for detecting at least one biomarker of Alzheimer’s disease, the method comprising, obtaining a biological sample from a human patient; and contacting the biological sample with an array comprising specific-binding molecules for the at least one biomarker and detecting binding between the at least one biomarker and the specific binding molecules. In one aspect the biomaker detected is a gene therapy target.

[0083] In a sixth embodiment the disclosure provides a kit comprising an array containing sequences of biomarkers from Table 1 or Table 2 for use in a human patient. In one aspect, the kit further contains reagents for RNA isolation and biomarkers for tuberous sclerosis genetic disorder. In a further aspect, the kit further advantageously comprises a container and a label or instructions for collection of a sample from a human, isolation of cells, inducement of cells to become pluripotent stem cells, growth of patient-specific neural organoids, isolation of RNA, execution of the array and calculation of gene expression change and prediction of concurrent or future disease risk. In one aspect, the biomarkers can include biomarkers listed in Table 2. In another aspect, biomarkers can comprise any markers or combination of markers in Tables 1 and 2 or variants thereof.

[0084] In a seventh embodiment, the disclosure provides a method for detecting one or a plurality of biomarkers from different human chromosomes associated with Alzheimer’s disease or Alzheimer's disease comorbidity susceptibility using data analytics that obviates the need for whole genome sequence analysis of patient genomes. In one aspect the methods are used to determine gene expression level changes that are used to identity clinically relevant symptoms and treatments, time of disease onset, and disease severity. In yet another aspect the neural organoids are used to identity novel biomarkers that serve as data input for development of algorithm techniques as predictive analytics. In a further aspect the algorithmic techniques include artificial intelligence, machine and deep learning as predictive analytics tools for identifying biomarkers for diagnostic, therapeutic target and drug development process for disease. Gene expression measured in Alzheimer’s disease can encode a variant of a biomarker alterations encoding a nucleic acid variant associated with Alzheimer’s disease. In one embodiment the nucleic acid encoding the variant is comprised of one or more missense variants, missense changes, or enriched gene pathways with common or rare variants.

[0085] In an alterative embodiment the method for predicting a risk for developing Alzheimer's disease in a human, comprising: collecting a biological sample; measuring biomarkers in the biological sample; and detecting measured biomarkers from the sample that are differentially expressed in humans with Alzheimer’s disease wherein the measured biomarkers comprise those biomarkers listed in Table 2.

[0086] In a further embodiment the measured biomarker is a nucleic acid encoding human biomarkers or variants listed as listed in Table 1. In one aspect a plurality of biomarkers comprising a diagnostic panel for predicting a risk for developing Alzheimer’s disease in a human, comprising biomarkers listed in Tables 1 and 2, or variants thereof. In one aspect of the embodiment a subset of marker can be used, wherein the subset comprises a plurality of biomarkers from 2 to 200, or 2-150, 2-100, 2-50, 2-25, 2-20, 2-15, 2- 10, or 2-5 genes.

[008η In yet an alternative embodiment the measured biomarker is a nucleic acid panel for predicting risk of Alzheimer's disease in humans. The genes encoding the biomarkens listed in Table 1 or variants thereof. Said panel can be provided according to the invention as an array of diagnostically relevant portions of one ora plurality of these genes, wherein the array can comprise any method for immobilizing, permanently or transiently, said diagnostically relevant portions of said one ora plurality of these genes, sufficient for the array to be interrogated and changes in gene expression detected and, if desired, quantified. In alternative embodiments the array comprises specific binding compounds for binding to the protein products of the one or a plurality of these genes. In yet further alternative embodiments, said specific binding compounds can bind to metabolic products of said protein products of the one or a plurality of these genes. In one aspect the presence of Alzheimer’s disease is detected by detection of one or a plurality of biomarkers as identified in Table 6 (Alzheimer's disease Diagnostic Biomarkers).

[0088] Another embodiment of the invention disclosed herein uses the neural organoids derived from the human patient in the non-diagnostic realm. The neural organoids express markers characteristic of a large variety of neurons and also include markers for astrocytic, oligodendritic, microglial, and vascular cells. The neural organoids form all the major regions of the brain including the retina, cortex, midbrain, brain stem, and the spinal cord in a single brain structure expressing greater than 98% of the genes known to be expressed in the human brain. Such characteristics enable the neural organoid to be used as a biological platform / device for drug screening, toxicity, safety, and/or pharmaceutical efficacy studies understood by those having skill in the art. Additionally, since the neural organoid is patient specific, pharmaceutical testing using the neural organoid allows for patient specific pharmacotherapy.

[00893 |n an eighth embodiment the disclosure provides methods for predicting a risk for developing Alzheimer's disease in a human, the method comprising procuring one or a plurality of cell samples from a human, comprising one ora plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more patient specific neural organoids; collecting a biological sample from the patient specific neural organoid; measuring biomarkers in the neural organoid sample; and detecting measured biomarkers from the neural organoid sample that are differentially expressed in humans with Alzheimer’s disease. (Clifford et al, Alzheimer’s & Dementia, 14; 535-562 (2018) "FDA floats new rules fortesting Alzheimer's drugs”. John Carrol, http://www.sciencemag.org/news/2018/02/fda-floats-new-rules- testing- alzheimers-drugs). In one aspect the one cell sample reprogrammed to the induced pluripotent stem cell is a fibroblast. In certain aspects the measured biomarkers comprise nucleic acids, proteins, or their metabolites. In further aspects, the measured biomarker is a nucleic acid encoding human A2M and APP-variant. In further aspects, the measured biomarkers comprise one or a plurality of genes as identified in Tables 1, 2, 5 or 6. In additional aspects , the neural organoid sample is procured from minutes to hours up to 15 weeks post inducement, wherein the the biomarkers to be tested are one or a plurality of biomarkers in Tables 5 or 6 (Diagnostic Neural Organoid Authentication Genes).

NEURAL ORGANOIDS AND EXOSOMES

[0090] Exosomes are extracellular vesicles that are released from cells upon fusion of the multivesicular body with the plasma membrane. The extracellular vesicles contain proteins and RNA packets containing micro and messenger RNAs that are transferred between cells. As such, the composition of the exosome reflects the origin cell. This property allows for the use of exosomes to predict disease onset, as well as novel therapeutic agents.

[0091] In one embodiment is a method for growing and isolating exosomes from healthy individuals. Such individuals are free from diseases including, but not limited to Alzheimer's disease, autism, Parkinson's disease, and cancer. The harvesting of exosomes from healthy individuals allows for the isolation of exosome-based RNA and proteins that serve as biomarkers and therapeutic agents for treating disease conditions such as Alzheimer's disease, autism, Parkinson's disease, and cancer. The embodiment comprises procurement of one or a plurality of cell samples from a healthy human, reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more therapeutic patient specific healthy neural organoids; and collecting exosomes, and exosome nucleic acids, proteins and metabolites from a plurality of the therapeutic, patient specific healthy neural organoid.

[0092] In a further embodiment is a method by which exosome RNA and proteins from healthy individuals are utilized in concert with exosome RNA and proteins isolated from a non-healthy individual at predefined time points, noted herein as scaled harvesting, to predict disease onset while also being therapeutic targets. The method comprises procuring one or a plurality of condition-specific samples from a sample including, but not limited to Alzheimer's disease, autism, Parkinson's disease, or cancer; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more condition-specific, patient specific, neural organoids; collecting exosome nucleic acid and protein from a plurality of the condition-specific patient specific neural organoids; detecting changes in the disease-specific exosome nucleic acids and proteins that are differentially expressed; performing assays on the condition-specific exosome nucleic acids and proteins to identify therapeutic agents that alter the differentially expressed in the condition specific versus healthy human exosome nucleic acids and protein profile; and administering a therapeutic agent to the individual.

[0093] The neural organoid of the current application is novel in that it allows for a scaled harvesting of exosomes at time points from minutes to hours to up to 15 weeks post inducement. The scaled harvesting of exosomes allows for identification of changes in exosome gene and protein biomarker expression patters that are indicative of disease onset. The presence of exosome gene and protein expression patterns indicative of disease onset subsequently can serve as therapeutic targets. Consistent with this, exosome nucleic acid and protein biomarkers from healthy individuals are harvested, fractionated, and/or enriched for specific biomarkers altered in the exosomes of Alzheimer's Disease, autism .Parkinson's disease, or cancer and used directly as therapeutic agents [0094] In one embodiment the exosomes can be collected at minutes to days after the neural organoid is generated. In a further embodiment, the exosome is isolated from the neural organoid and the nucleic acids and proteins harvested up to 15 weeks after induction of the neural organoid.

[0095] In a further embodiment, exosomes can be isolated at minutes, hours, days, or weeks after reprogramming. For instance, exosomes can be harvested at about 10 minutes,

20 minutes, 30 minutes, 40 minutes, 50 minutes, and 60 minutes. In a further embodiment the exosomes can be harvested 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, or 24 hours. In yet a further embodiment the exosome can be harvested at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, or 1 week, 2 weeks, 3 weeks, 4 weeks,

5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks 10 weeks, 11 weeks, 12 weeks or more in culture.

[0096] Exosomes collected at a wide range of time points, referred to as scaled harvesting herein, allow for insights and data related to regulatory RNA changes that are indicative of disease onset. In one embodiment, the scaled harvesting allows for enrichment of specific biomarkers collected at specific time points from the normal human exosome. Moreover, exosomes can be fractionated and/or enriched to increase yields or enhance therapeutic and predictive responses.

[0097] The numerous time points are invaluable in predicting disease occurrence / onset and also provide a novel mechanism for therapeutic agents in numerous conditions, including but not limited to Alzheimer's disease, Parkinson’s Disease, malignant and cancerous tumors, autism, and associated co-morbidities. In one embodiment the neural organoid can be used to establish an exosome profile database (See APL Bioeng. 2019 Mar; 3(1)) that can be utilized for determining biomarkers characteristic of disease onset and timing of disease onset. In another embodiment the effectiveness of treatment strategies and therapeutic agents for a wide range of conditions can be evaluated, based on changes in neuronal organoid derived exosomes.

[0098] In yet another embodiment, the nucleic acids and proteins isolated from the exosome of the neural organoid from the healthy human are utilized to construct a biomarker library and evaluate disease onset and predict disease risk.

[0099] In yet another embodiment, the alterations in exosome RNA and protein expression can be used to predict the risk of developing Alzheimer's disease, autism, Parkinson's disease, or cancer or tumor in a human. In an intial step the exosome from a healthy individual is isolated, more specifcally, the method comprises; procuring one or a plurality of cell samples from a healthy human, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more therapeutic patient specific healthy neural organoids; and collecting exosome nucleic acids and proteins from a plurality of the therapeutic, patient specific healthy neural organoid.

[00100] The method further comprises procuring one or a plurality of cell samples from a human with Alzheimer's disease, autism, Parkinson's disease, or cancer or tumor, comprising one or a plurality of cell types; reprogramming the one or the plurality of cell samples to produce one or a plurality of induced pluripotent stem cell samples; treating the one or the plurality of induced pluripotent stem cell samples to obtain one or more Alzheimer's disease, autism, Parkinson's disease, or cancer or tumor patient specific, neural organoids; collecting exosome nucleic acid and protein from the patient specific neural organoids; detecting changes in Alzheimer's disease, autism, Parkinson's disease, or cancer or tumor disease exosome nucleic acid and proteins that are differentially expressed in humans with the condition; performing assays on the Alzheimer's disease, autism, Parkinson's disease, or cancer or tumor disease exosome nucleic acids and proteins to identify therapeutic agents that alter the differentially expressed exosome nucleic acids and protein; and administering a therapeutic agent to the human. [00101] The exosome biomarkers used in the prediction and treatment of a condition comprise nucleic acids, proteins, or their metabolites and may include A2M, APR, and associated variants. The biomarkers may further comprise one or a plurality of genes as identified in Tables 1 , 2, 5 or 6.

[00102] In a further embodiment neural organoids can be used to identify novel biomarkers that serve as data input for development of algorithm techniques such artificial intelligence, machine and deep learning, including biomarkers for diagnostic, therapeutic target and drug development process for disease. The use of data analytics for relevant biomarker analysis permits detection of autism and comorbidity susceptibility, thereby obviating the need for whole genome sequence analysis of patient genomes.These and other data findings, features, and advantage of the present disclosure will be more fully understood from the detailed description taken together with the accompanying claims. It is noted that the scope of the claims is defined by the recitations therein and not by the specific discussion of features and advantages set forth in the present description.

[00103] These and other data findings, features, and advantage of the present disclosure will be more fully understood from the detailed description taken together with the accompanying claims. It is noted that the scope of the claims is defined by the recitations therein and not by the specific discussion of features and advantages set forth in the present description

EXAMPLES

[00104] The Examples that follow are illustrative of specific embodiments of the invention, and the use thereof. It is set forth for explanatory purposes only and is not taken as limiting the invention. In particular, the example demonstrates the effectiveness of neural organoids in predicting future disease risk.

[00105]

MATERIALS AND METHODS

[00106] The neural organoids described above were developed using the following materials and methods.

Summary of Methods:

[00107] Neural Organoids derived from induced pluripotent stem cells derived from adult skin cells of patients were grown in vitro for 4 weeks as previous described in our PCT Application (PCT/US2017/013231). Transcriptomic data from these neural organoids were obtained. Differences in expression of 20,814 genes expressed in the human genome were determined between these neural organoids and those from neural organoids from a normal individual human. Detailed data analysis using Gene Card and Pubmed data bases were performed. Genes that were expressed at greater than 1.4 fold were found to be highly significant because a vast majority were correlated with genes previously associated with a multitude of neurodevelopmental and neurodegenerative diseases as well as those found to be dysregulated in post mortem patient brains. These genes comprise a suite of biomarkers for Alzheimer’s disease.

[00108] The invention advantageously provides many uses, including but not limited to a) early diagnosis of these diseases at birth from new bom skin cells; b) Identification of biochemical pathways that increase environmental and nutritional deficiencies in new bom infants; c) discovery of mechanisms of disease mechanisms; d) discovery of novel and early therapeutic targets for drug discovery using timed developmental profiles; e) testing of safety, efficacy and toxicity of drugs in these pre-clinical models.

[00109] Cells used in these methods include human iPSCs, feeder-dependent (System Bioscience. Wf SC600A-W) and CF-1 mouse embryonic fibroblast feeder cells, gamma- irradiated (Applied StemCell, Inc #ASF- 1217)

[00110] Growth media, or DMEM media, used in the examples contained the supplements as provided in Table 3 (Growth Media and Supplements used in Examples).

Table 3: Growth Media and Supplements used in Examples

[00111] One skilled in the art will recognize that additional formulations of media and supplements can be used to culture, induce and maintain pluripotent stem cells and neural organoids.

[00112J Experimental protocols required the use of multiple media compositions including MEF Media, IPSC Media, EB Media, Neural Induction Media, and Differentiation Medias 1,

2, and 3.

[00113] Mouse embryonic fibroblast (MEF) was used in cell culture experiments. MEF Media comprised DMEM media supplemented with 10% Feta Bovine Serum, 100 units/ml penicillin, 100 microgram/ml streptomycin, and 0.25 microgram/ml Fungizone.

[00114] Induction media for pluripotent stem cells (IPSC Media) comprised DMEM/F12 media supplemented with 20% Knockout Replacement Serum, 3% Fetal Bovine Serum with 2mM Glutamax, IX Minimal Essential Medium Nonessential Amino Acids, and 20 nanogram/ml basic Fibroblast Growth Factor

[00115] Embryoid Body (EB) Media comprised Dulbecco's Modified Eagle's Medium (DMEM) (DMEM)/Ham's F-12 media, supplemented with 20% Knockout Replacement Serum, 3% Fetal Bovine Serum containing 2mM Glutamax, IX Minimal Essential Medium containing Nonessential Amino Acids, 55microM beta-mercaptoethanol, and 4ng/ml basic Fibroblast Growth Factor.

[00116] Neural Induction Media contained DMEM/F12 media supplemented with: a 1 :50 dilution N2 Supplement, a 1 :50 dilution GlutaMax, a 1:50 dilution MEM-NEAA, and 10 microgram/ml Heparin '

[00117] Three differentiation medias were used to produce and grow neural organoids. Differentiation Media 1 contained DMEM/F12 media and Neurobasal media in a 1:1 dilution. Each media is commercially available from Invitrogen. The base media was supplemented with a 1 :200 dilution N2 supplement, a 1 :100 dilution B27 - vitamin A, 2.5microgram/ml insulin, 55microM beta-mercaptoethanol kept under nitrogen mask and frozen at -20 e C, 100 units/ml penicillin, 100 microgram/ml streptomycin, and 0.25microgram/ml Fungizone.

[00118] Differentiation Media 2 contained DMEM/F12 media and Neurobasal media in a 1:1 dilution supplemented with a 1:200 dilution N2 supplement, a 1:100 dilution B27 containing vitamin A, 2.5microgram/ml Insulin, 55umicroMolar beta-mercaptoethanol kept under nitrogen mask and frozen at -20°C, 100units/ml penicillin, 100microgram/ml streptomycin, and 0.25microgram/ml Fungizone.

[00119] Differentiation Media 3 consisted of DMEM/F12 media: Neurobasal media in a 1:1 dilution supplemented with 1:200 dilution N2 supplement, a 1:100 dilution B27 containing vitamin A), 2.5microgram/ml insulin, 55microMolar beta-mercaptoethanol kept under nitrogen mask and frozen at -20°C, 100 units/ml penicillin, 100 microgram/ml streptomycin, 0.25microgram/ml Fungizone, TSH, and Melatonin.

[00120] The equipment used in obtaining, culturing and inducing differentiation of pluripotent stem cells is provided in Table 4 (Equipment used in Experimental Procedures). One skilled in the art would recognize that the list is not at all exhaustive but merely exemplary.

Table 4: Equipment used in Experimental Procedures.

Example 1: Generation of human induced pluripotent stem cell-derived neural organoids. [00121] Human induced pluripotent stem cell-derived neural organoids were generated according to the following protocol, as set forth in International Application No.

PCT/US2017/013231 incorporated herein by reference. Briefly, irradiated murine embryonic fibroblasts (MEF) were plated on a gelatin coated substrate in MEF media (Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% Feta Bovine Serum, 100 units/ml penicillin, 100 microgram/ml streptomycin, and 0.25 microgram/ml Fungizone) at a density of 2 x 10 s cells per well. The seeded plate was incubated at 37°C overnight. [00122] After incubation, the MEFs were washed with pre-warmed sterile phosphate buffered saline (PBS). The MEF media was replaced with 1 mL per well of induced pluripotent stem cell (iPSC) media containing Rho-associated protein kinase (ROCK) inhibitor. A culture plate with iPSCs was incubated at 37°C. The iPSCs were fed every other day with fresh iPSC media containing ROCK inhibitor. The iPSC colonies were lifted, divided, and transferred to the culture wells containing the MEF cultures so that the iPSC and MEF cells were present therein at a 1 :1 ratio. Embryoid bodies (EB) were then prepared. Briefly, a 100 mm culture dish was coated with 0.1% gelatin and the dish placed in a 37°C incubator for 20 minutes, after which the gelatin-coated dish was allowed to air dry in a biological safety cabinet. The wells containing iPSCs and MEFs were washed with prewarmed PBS lacking Ca 2+/ Mg 2+ . A pre-warmed cell detachment solution of proteolytic and collagenolytic enzymes (1 mL/well) was added to the iPSC/MEF cells. The culture dishes were incubated at 37°C for 20 minutes until cells detached. Following detachment, pre- warmed iPSC media was added to each well and gentle agitation used to break up visible colonies. Cells and media were collected and additional pre-warmed media added, bringing the total volume to 15 mL. Cells were placed on a gelatin-coated culture plate at 37°C and incubated for 60 minutes, thereby allowing MEFs to adhere to the coated surface. The iPSCs present in the cell suspension were then counted.

[00123] The suspension was then centrifuged at 300xg for 5 minutes at room temperature, the supernatant discarded, and cells re-suspended in EB media supplemented with ROCK inhibitor (50uM final concentration) and 4ng/ml basic Fibroblast Growth Factor to a volume of 9,000 cells/150 μL. EB media is a mixture of DMEM/Ham's F-12 media supplemented with 20% Knockout Replacement Serum, 3% Fetal Bovine Serum (2mM Glutamax), 1X Minimal Essential Medium Nonessential Amino Acids, and 55 μΜ beta- mercaptoethanol. The suspended cells were plated (150 μL) in a LIPIDURE® low- attachment U-bottom 96-well plate and incubated at 37°C.

[00124] The plated cells were fed every other day during formation of the embryoid bodies by gently replacing three fourths of the embryoid body media without disturbing the embryoid bodies forming at the bottom of the well. Special care was taken in handling the embryoid bodies so as not to perturb the interactions among the iPSC cells within the EB through shear stress during pipetting. For the first four days of culture, the EB media was supplemented with 50uM ROCK inhibitor and 4ng/ml bFGF. During the remaining two to three days the embryoid bodies were cultured, no ROCK inhibitor or bFGF was added. [00125] On the sixth or seventh day of culture, the embryoid bodies were removed from the LIPIDURE® 96 well plate and transferred to two 24-well plates containing 500 μL/well Neural Induction media, DMEM/F12 media supplemented with a 1:50 dilution N2 Supplement, a 1 :50 dilution GlutaMax, a 1 :50 dilution MEM-Non-Essential Amino Acids (NEAA), and 10 pg/ml Heparin. Two embryoid bodies were plated in each well and incubated at 37°C. The media was changed after two days of incubation. Embryoid bodies with a "halo" around their perimeter indicate neuroectodermal differentiation. Only embryoid bodies having a "halo" were selected for embedding in matrigel, remaining embryoid bodies were discarded.

[00126] Plastic paraffin film (PARAFILM) rectangles (having dimensions of 5cm x 7cm) were sterilized with 3% hydrogen peroxide to create a series of dimples in the rectangles. This dimpling was achieved, in one method, by centering the rectangles onto an empty sterile 200μL tip box press, and pressing the rectangles gently to dimple it with the impression of the holes in the box. The boxes were sprayed with ethanol and left to dry in the biological safety cabinet.

[00127] Frozen Matrigel matrix aliquots (500 μL) were thawed on ice until equilibrated at 4 e C. A single embryoid body was transferred to each dimple of the film. A single 7cm x 5cm rectangle holds approximately twenty (20) embryoid bodies. Twenty microliter (20μL) aliquots of Matrigel were transferred onto the embryoid bodies after removing extra media from the embryoid body with a pipette. The Matrigel was incubated at 37°C for 30 min until the Matrigel polymerized. The 20μL droplet of viscous Matrigel was found to form an optimal three dimensional environment that supported the proper growth of the neural organoid from embryoid bodies by sequestering the gradients of morphogens and growth factors secreted by cells within the embryoid bodies during early developmental process. However, the Matrigel environment permitted exchange of essential nutrients and gases. Gentle oscillation by hand twice a day for a few minutes within a tissue culture incubator (37°C/5%C02) further allowed optimal exchange of gases and nutrients to the embedded embryoid bodies.

[00128] Differentiation Media 1 , a one-to-one mixture of DMEM/F12 and Neurobasal media supplemented with a 1:200 dilution N2 supplement, a 1:100 dilution B27 - vitamin A, 2.5 pg/mL insulin, 55 microM beta-mercaptoethanol kept under nitrogen mask and frozen at -20°C, 100 units/mL penicillin, 100 pg/mL streptomycin, and 0.25 pg/mL Fungizone, was added to a 100 mm tissue culture dish. The film containing the embryoid bodies in Matrigel was inverted onto the 100 mm dish with differentiation media 1 and incubated at 37 e C for 16 hours. After incubation, the embryoid body/Matrigel droplets were transferred from the film to the culture dishes containing media. Static culture at 37 e C was continued for 4 days until stable neural organoids formed.

[00129] Organoids were gently transferred to culture flasks containing differentiation media 2, a one-to-one mixture of DMEM/F12 and Neurobasal media supplemented with a 1 :200 dilution N2 supplement, a 1 :100 dilution B27 + vitamin A, 2.5 pg/mL insulin, 55microM beta-mercaptoethanol kept under nitrogen mask and frozen at -20°C, 100 units/mL penicillin, 100 pg/mL streptomycin, and 0.25 pg/mL Fungizone. The flasks were placed on an orbital shaker rotating at 40 rpm within the 37°C/5% CO2 incubator.

[00130] The media was changed in the flasks every 3-4 days to provide sufficient time for morphogen and growth factor gradients to act on targets within the recipient cells forming relevant structures of the brains. Great care was taken when changing media so as to avoid unnecessary perturbations to the morphogen/secreted growth factor gradients developed in the outer most periphery of the organoids as the structures grew into larger organoids. [00131] FIG.16 illustrates neural organoid development in vitro. Based on transcriptomic analysis, iPSC cells form a body of cells alter 3D culture, which become neural progenitor cells (NPC) after neural differentiation media treatment. Neurons were observed in the cell culture after about one week. After about four (4) weeks or before, neurons of multiple lineage appeared. At about twelve (12) weeks or before, the organoid developed to a stage having different types of cells, including microglia, oligodendrocyte, astrocyte, neural precursor, neurons, and intemeurons.

Example 2: Human induced pluripotent stem cell-derived neural organoids express characteristics of human brain development.

[00132] After approximately 12 weeks of in vitro culture, transcriptomic and immunohistochemical analysis indicated that organoids were generated according to the methods delineated in Example 1. Specifically, the organoids contained cells expressing markers characteristic of neurons, astrocytes, oligodendrocytes, microglia, and vasculature (FIGs. 1-14) and all major brain structures of neuroectodermal derivation. Morphologically identified by bright field imaging, the organoids included readily identifiable neural structures including cerebral cortex, cephalic flexure, and optic stalk ( compare , Grey's Anatomy Textbook). The gene expression pattern in the neural organoid was >98 % concordant with those of the adult human brain reference (Clontech, #636530). The organoids also expressed genes in a developmental^ organized manner described previously (e.g. for the midbrain mesencephalic dopaminergic neurons; Blaese et al., Genetic control of midbrain dopaminergic neuron development. Rev Dev Biol. 4(2): 113-34, 2015). The structures also stained positive for multiple neural specific markers (dendrites, axons, nuclei), cortical neurons (Doublecortin), midbrain dopamine neurons (Tyrosine Hydroxylase), and astrocytes (GFAP) as shown by immunohistology).

[00133] All human neural organoids were derived from iPSCs of fibroblast origin (from System Biosciences, Inc). The development of a variety of brain structures was characterized in the organoids. Retinal markers are shown in FIG.15. Doublecortin (DCX), a microtubule associated protein expressed during cortical development, was observed in the human neural organoid (FIG. 1A and FIG. 1B, and FIG. 16). Midbrain development was characterized by the presence of tyrosine hydroxylase (FIG. 2). In addition, transcriptomics revealed expression of the midbrain markers DLKI, KLHL I, and PTPRU (FIG. 6A). GFAP staining was used to identify the presence of astrocytes in the organoids (FIG. 3). NeuN positive staining indicated the presence of mature neurons (FIG. 3). In addition, the presence of NKCCI and KCC2, neuron-specific membrane proteins, was observed in the organoid (FIG. 4). A schematic of the roles of NKCCI and KCC2 is provided in FIG. 5A. FIG. 5B indicates that a variety of markers expressed during human brain development are also expressed in the organoids described in Example 1.

[00134] Markers expressed within the organoids were consistent with the presence of excitatory, inhibitory, cholinergic, dopaminergic, serotonergic, astrocytic, oligodendritic, microglial, vasculature cell types. Further, the markers were consistent with those identified by the Human Brain Reference (HBR) from Clontech (FIG. 5C) and were reproducible in independent experiments (FIG. 5D). Non-brain tissue markers were not observed in the neural organoid (FIG. 6B).

[00135] Tyrosine hydroxylase, an enzyme used in the synthesis of dopamine, was observed in the organoids using immunocytochemistry (FIG. 5B) and transcriptomics (FIG. 6A). The expression of other dopaminergic markers, including vesicular monoamine transporter 2 (VMAT2), dopamine active transporter (DAT) and dopamine receptor D2 (D2R) were observed using transcriptomic analysis. FIG. 7 delineates the expression of markers characteristic of cerebellar development. FIG. 8 provides a list of markers identified using transcriptomics that are characteristic of neurons present in the hippocampus dentate gyrus. Markers characteristic of the spinal cord were observed after 12 weeks of in vitro culture. FIG. 9 provides a list of markers identified using transcriptomics that are characteristic of GABAergic intemeuron development. FIG. 10 provides a list of markers identified using transcriptomics that are characteristic of the brain stem, in particular, markers associated with the serotonergic raphe nucleus of the pons. FIG. 11 lists the expression of various Hox genes that are expressed during the development of the cervical, thoracic and lumbar regions of the spinal cord.

[00136] FIG. 12 shows that results are reproducible between experiments. The expression of markers detected using transcriptomics is summarized in FIG. 13.

[00137] In sum, the results reported herein support the conclusion that the invention provides an in vitro cultured organoid that resembles an approximately 5 week old human fetal brain, based on size and specific morphological features with great likeness to the optical stock, the cerebral hemisphere, and cephalic flexure in a 2-3mm organoid that can be grown in culture. High resolution morphology analysis was carried out using immunohistological methods on sections and confocal imaging of the organoid to establish the presence of neurons, axons, dendrites, laminar development of cortex, and the presence of midbrain marker.

[00138] This organoid includes an interactive milieu of brain circuits as represented by the laminar organization of the cortical structures in Fig. 13 and thus supports formation of native neural niches in which exchange of miRNA and proteins by exosomes can occur among different cell types.

[00139] Neural organoids were evaluated at weeks 1 , 4 and 12 by transcriptomics. The organoid was reproducible and replicable (FIGs. 5C, 5D, FIG. 12, and FIG. 18). Brain organoids generated in two independent experiments and subjected to transcriptomic analysis showed >99% replicability of the expression pattern and comparable expression levels of most genes with <2-fold variance among some of the replicates.

[00140] Gene expression patterns were analyzed using whole genome transcriptomics as a function of time in culture. Results reported herein indicate that within the neural organoid known developmental order of gene expression in vivo occurs, but on a somewhat slower timeline. For example, the in vitro temporal expression of the transcription factors NURRI and PITX3, genes uniquely expressed during midbrain development, replicated known in vivo gene expression patterns (Fig 6A). Similarly, the transition from GABA mediating excitation to inhibition, occurred following the switch of the expression of the Na(*)-K(*)-2CI(- -)) cotransporter NKCCI (SLC12A2), which increases intracellular chloride ions, to the K(*)- Cl( ) cotransporter KCC2 (SLC12A5) (Owens and Kriegstein, Is there more to GABA than synaptic inhibition?, Nat Rev Neurosd. 3(9):715-272002), which decreases intracellular chloride ion concentrations (Blaesse et al., Cation-chloride cotransporters and neuronal function. Neuron. 61(6) 820-838, 2009). Data on the development of the brain organoids in culture showed that expression profiles of NKCCI and KCC2 changed in a manner consistent with an embryonic brain transitioning from GABA being excitatory to inhibitory (Fig. 4 & 5), a change that can be monitored by developmental transcriptomics.

Example 3: Tuberous sclerosis complex model

[00141] Tuberous sclerosis complex (TSC) is a genetic disorder that causes non- malignant tumors to form in multiple organs, including the brain. TSC negatively affects quality of life, with patients experiencing seizures, developmental delay, intellectual disability, gastrointestinal distress and Alzheimer’s disease. Two genes are associated with TSC: (1) the TSC1 gene, located on chromosome 9 and also referred to as the hamartin gene and (2) the TSC2 gene located on chromosome 16 and referred to as the tuberin gene.

[00142] Using methods as set forth in Example 1 , a human neural organoid from iPSCs was derived from a patient with a gene variant of the TSC2 gene (ARG I743GLN) from iPSCs (Cat# GM25318 Coriell Institute Repository, NJ). This organoid served as a genetic model of a TSC2 mutant.

[00143] Both normal and TSC2 mutant models were subject to genome-wide transcriptomic analysis using the Ampliseq™ analysis (ThermoFisher) to assess changes in gene expression and how well changes correlated with the known TSC clinical pathology (Fig. 14).

[00144] Whole genome transcriptomic data showed that of all the genes expressed (13,000), less than a dozen showed greater than two-fold variance in the replicates for both Normal N)) and TSC2. This data supported the robustness and replicability of the human neural organoids at week 1 in culture.

[00145] Clinically TSC patients present with tumors in multiple organs including the brain, lungs, heart, kidneys and skin (Harmatomas). In comparison of WT and TSC2, the genes expressed at two-fold to 300-fold differences, included those correlated with 1) tumor formation and 2) Alzheimer's disease mapped using whole genome and exome sequencing strategies. 2

Example 4: Human Neural Organoid Model Gene Expression to Predict Alzheimer’s disease

[00146] Alzheimer's disease and Alzheimer’s disease spectrum disorder is a development disorder that negatively impacts social interactions and day-to-day activities. In some cases, the disease can include repetitive and unusual behaviors and reduced tolerance for sensory stimulation. Many of the Alzheimer’s disease-predictive genes are associated with brain development, growth, and/or organization of neurons and synapses. [00147] Alzheimer's disease has a strong genetic link with DNA mutations comprising a common molecular characteristic of Alzheimer’s disease. Alzheimer’s disease encompasses a wide range of genetic changes, most often genetic mutations. The genes commonly identified as playing a role in Alzheimer’s disease include novel markers provided in Table 1 and Alzheimer's disease markers provided in Table 2.

[00148] Expression changes and mutations in the noted genes disclosed herein from the neural organoid at about week 1 , about week 4 and about week 12 are used in one embodiment to predict future Alzheimer’s disease risk. In a further aspect, mutations in the genes disclosed can be determined at hours, days or weeks after reprogramming.

[00149] In a second embodiment, mutations in Table 1 , in the human neural organoid at about week 1 , about week 4, and about week 12 are used to predict the future risk of Alzheimer’s disease using above described methods for calculating risk. One skilled in the art would recognize that additional biomarker combinations expressed in the human neural organoid can also be used to predict future Alzheimer’s disease onset. [00150] The model used herein is validated and novel in that data findings reconcile that the model expresses four hundred and seventy two markers of Alzheimer’s disease patient post mortem brains and databases (Table 2), as shown in Table 5. The model is novel in that it uses, as starting material, an individual’s iPSCs originating from skin or blood cells as the starting material to develop a neural organoid that allows for identification of Alzheimer’s disease markers early in development including at birth

Table 5. Therapeutic Neural Organoid Authentication Genes

[00151] One of skill in the art will recognize that sequence data for the genes listed above can be obtained in publicly available gene databases such as GeneCards, GenBank, Malcard, Uniport and PathCard databases.

Table 6. Diagnostic Neural Organoid Authentication Genes UNC13A

VAMP2

VAV3

VCAN

VSNL1

VWA5B1

WASF1

WDR16

WDR17

WDR47

WDR63

WDR96

WIF1

ZBTB16

ZDBF2

ZFHX3

ZNF804A

[00152] One of skill in the art will recognize that sequence data for the genes listed above can be obtained in publicly available gene databases such as GeneCands, GenBank, Malcard, Uniport and PathCard databases.

Example 5: Predicting Risk of Disease Onset from Neural Organoid Gene Expression [00153] Gene expression in the neural organoid can be used to predict disease onset. Briefly, gene expression is correlated with Gene Card and Pubmed database genes and expression compared for dysregulated expression in diseased vs non-disease neural organoid gene expression.

Example 6: Prediction of Co-Morbidities Associated with Alzheimer’s disease [00154] The human neural organoid model data findings can be used in the prediction of comorbidity onset or risk associated with Alzheimer’s disease including at birth (Reference: European Bioinformatic Institute (EBI) and ALLEN INSTITUTE databases) and in detecting comorbidities, genes associated with one or more of these diseases are detected from the patient’s grown neural organoid. Such genes include, comorbidities and related accession numbers include, those listed in Table 7:

Table 7: Genes and Co-Morbidity Susceptibility/Resistance Associated with Alzheimer’s Disease

Gene AD Comorbidity SusceptibilHy/Resistance (Reft GeneCards)

ABCA4 Macular Degeneration, Age-Related, 2 and Starganclt Disease 1.

ABCB1 Colchicine Resistance and Inflammatory Bowel Disease 13.

ABCB11 Cholestasis, Progressive Familial Intrahepatic, 2 and Cholestasis, Benign Recurrent Intrahepatic, 2.

ABCC5 Lymphoblastic Leukemia.

ABCC6 Pseudoxanthoma Elasticum and Arterial Calcification, Generalized, Of Infancy, 2.

ABCC8 Hyperinsulinemic Hypoglycemia, Familial, 1 and Hypoglycemia, Leucine- Induced.

ABCD2 Adrenoleukodystrophy and Demyelinating Disease.

ACACB Biotin Deficiency and Diabetes Mellitus, Noninsulin-Dependent.

ASIC3 Frozen Shoulder and Deafness, Autosomal Recessive 13.

ACOT7 Raynaud Disease and Meckel Diverticulum.

ACR Spermatogenic Failure 6 and Male Infertility.

ACSL6 Myelodysplastic Syndromeand Chronic Intestinal Vascular Insufficiency.

ACSM3 Pneumothorax, Primary Spontaneous.

ACTG2 Visceral Myopathy and Chronic Intestinal Pseudoobstruction.

ACTN2 Cardiomyopathy, Dilated, 1Aa, With Or Without Left Ventricular Noncompaction and Atrial Standstill 1.

ACTRT1 Bazex Syndrome.

ADAM22 Epileptic Encephalopathy, Early Infantile, 61 and Brachydactyly, Type C.

ADAM23 Developmental Biology and LGI-ADAM interactions.

ADAMTS2 Ehlers-Danlos Syndrome, Dermatosparaxis Type and Ehlers-Danlos Syndrome.

ADAMTS3 Hennekam Lymphangiectasia-Lymphedema Syndrome 3 and Hennekam Syndrome.

ADAMTS8 Peters-Plus Syndrome.

ADARB1 Dyschromatosis Symmetrica Hereditaria and Aik-Negative Anaplastic Large Cell Lymphoma. ADD2 Hereditary Elliptocytosis and Capillariasis.

AFF2 Mental Retardation, X-Linked, Associated With Fragile Site Fraxe and Fragile X Syndrome.

AGT Renal Tubular Dysgenesis and Hypertension, Essential.

AHNAK2 Hard Palate Cancer.

AK5 Anterograde Amnesia and Prosopagnosia.

AK7 Spenmatogenic Failure 27 and Non-Syndromic Male Infertility Due To Sperm Motility Disorder.

AKR1B10 Tobacco Addiction.

AKR1C2 46, Xy Sex Reversal 8 and Perrault Syndrome 1.

ALDH1A1 Lung Adenoma and Erythroplakia.

ALOX5AP Stroke, Ischemic and Macular Holes.

AMHR2 Persistent Mullerian Duct Syndrome, Types I And li and Persistent Mullerian Duct Syndrome.

AMPD3 Erythrocyte Amp Deaminase Deficiency and Adenosine Monophosphate Deaminase 1 Deficiency.

ANK1 Spherocytosis, Type 1 and Hereditary Spherocytosis.

ANKRD37 Low Density Lipoprotein Receptor-Related Protein Binding Protein

ANLN Focal Segmental Glomerulosclerosis 8 and Familial Idiopathic Steroid- Resistant Nephrotic Syndrome With Focal Segmental Hyalinosis.

AN05 Gnathodiaphyseal Dysplasia and Miyoshi Muscular Dystrophy 3.

AP3B2 Epileptic Encephalopathy, Early Infantile, 48 and Undetermined Early- Onset Epileptic Encephalopathy.

APBB2 Perrault Syndrome 1 and Alzheimer Disease.

APOD Breast Cyst and Niemann-Pick Disease.

APOL4 Schizophrenia.

AREG Colorectal Cancer and Psoriasis.

ARHGAP18 Lice Infestation and Penicilliosis.

ARHGAP31 Adams-Oliver Syndrome 1 and Adams-Oliver Syndrome.

ARHGEF9 Epileptic Encephalopathy, Early Infantile, 8 and Hyperekplexia.

ARMC4 Ciliary Dyskinesia, Primary, 23 and Kartagener Syndrome.

ARSI Autosomal Recessive Spastic Paraplegia Type 66 and Louse-Borne Relapsing Fever.

ASPN Intervertebral Disc Disease and Osteoarthritis. ASRGL1 Telogen Effluvium and Mass Syndrome.

ASTN2 Bartiet-Biedl Syndrome 11 and Migraine Without Aura.

ATOH7 Persistent Hyperplastic Primary Vitreous, Autosomal Recessive and Persistent Hyperplastic Primary Vitreous.

ATP1A3 Parkinson's

ATP2B2 Deafness, Autosomal Recessive 12 and Chromosome 3Pter-P25 Deletion Syndrome.

ATP2B3 Spinocerebellar Ataxia, X-Linked 1 and Muscular Atrophy.

ATP8A2 Cerebellar Ataxia, Mental Retardation, And Dysequilibrium Syndrome 4 and Cerebellar Ataxia, Mental Retardation, And Dysequilibrium Syndrome 1.

B4GALNT1 Spastic Paraplegia 26, Autosomal Recessive and Spastic Paraplegia 26.

BACH2 Schuurs-Hoeijmakers Syndrome and Smoldering Myeloma.

BHLHE22 Mental Retardation, X-Linked, Syndromic, Martin-Probst Type and Phosphoglycerate Dehydrogenase Deficiency.

BOC Leber Congenital Amaurosis 4.

BRSK2 Limbic Encephalitis and Pleural Tuberculosis.

BSN Decubitus Ulcer and Chronic Ulcer Of Skin.

BST2 Stomatitis and West Nile Encephalitis.

BTC Cardiomyopathy, Familial Hypertrophic, 1.

C15orf26 Primary Ciliary Dyskinesia.

C1QB C1q Deficiency and Immunodeficiency Due To A Classical Component Pathway Complement Deficiency.

C1QC C1q Deficiency and Immunodeficiency Due To A Classical Component Pathway Complement Deficiency.

C20orf160 Cavernous Malformation and Cerebral Cavernous Malformations.

C20orf85 Lung Cancer.

C3AR1 Occupational Dermatitis and Complement Component 3 Deficiency.

C3orf35 Muir-Torre Syndrome.

SOGA3 Heart Conduction Disease.

FRRS1L Epileptic Encephalopathy, Early Infantile, 37and Chorea, Childhood- Onset, With Psychomotor Retardation.

CA10 Non-Suppurative Otitis Media and Chondroblastoma.

CABYR Suppurative Thyroiditis. CACNA1E NFAT and Cardiac Hypertrophy

CACNB1 Headache and Malignant Hyperthermia.

CACNB4 Episodic Ataxia, Type 5 and Epilepsy, Idiopathic Generalized 9.

CACNG2 Mental Retardation, Autosomal Dominant 10 and Autosomal Dominant Non-Syndromic Intellectual Disability.

CACNG4 Cardiac Hypertrophy and Fc-GammaR Pathway.

CACNG8 Dilated Cardiomyopathy.

CADM3 Cleft Lip/Palate-Ectodermal Dysplasia Syndrome.

CALB1 Huntington Disease and Temporal Lobe Epilepsy.

CALML4 Neuronal Ceroid Lipofuscinosis.

CALY Attention Deficit-Hyperactivity Disorder.

CAMK2B Mental Retardation, Autosomal Dominant 54 and Autosomal Dominant Non-Syndromic Intellectual Disability.

CAMTA1 Cerebellar Ataxia, Nonprogressive, With Mental Retardationand Epithelioid Hemangioendothelioma.

CAPN14 Esophageal Disease.

CAPN6 Leiomyosarcoma and Corneal Dystrophy, Posterior Polymorphous, 1.

CASP1 Cowpox and Shigellosis.

CASP6 Dystrophinopathies.

CASZ1 Retroperitoneal Sarcoma and Retrope ritoneum Carcinoma.

CBLN1 Depression.

CCDC103 Ciliary Dyskinesia, Primary, 17 and Ciliary Dyskinesia, Primary, 1.

CCDC19 Nasopharyngeal Disease and Pharynx Cancer.

CCDC65 Ciliary Dyskinesia, Primary, 27 and Primary Ciliary Dyskinesia.

CCIN Pelvic Varices.

CCL18 Gaucher's Disease and Pulmonary Fibrosis.

CCL3 Human Immunodeficiency Virus Type 1

CCL4 Pulmonary Tuberculosis and Meningitis.

CCP110 Spinocerebellar Ataxia 11 and Townes-Brocks Syndrome.

CD101 Langerhans Cell Histiocytosis and Histiocytosis.

CD109 Fetal And Neonatal Alloimmune Thrombocytopenia and Vulva Squamous Cell Carcinoma.

CD14 Mycobacterium Chelonae and Croup. CD163 Rosai-Dorfman Disease and Non-Langerhans-Cell Histiocytosis.

CD1C Mycobacterium Malmoense and Foramen Magnum Meningioma.

CD34 Dermatofibrosarcoma Protuberans and Gastrointestinal Stromal Tumor.

CD4 Okt4 Epitope Deficiency and Pilonidal Sinus.

CD68 Granular Cell Tumor and Breast Granular Cell Tumor.

CD? Pityriasis Lichenoides Et Varioliformis Acuta and T-Cell Leukemia.

CD74 Undifferentiated Pleomorphic Sarcoma and Mantle Cell Lymphoma.

CDC25C Plague and Prostate Cancer.

CDCA5 Cornelia De Lange Syndrome.

CDCA7L Medulloblastoma.

CDCP1 Colorectal Cancer.

CDH15 Autosomal Dominant Non-Syndromic Intellectual

Disability and Hypotrichosis, Congenital, With Juvenile Macular

Dystrophy.

CDH8 Learning Disability and Autism Spectrum Disorder.

CD01 Hepatoblastoma and Esophagus Adenocarcinoma.

CDX2 Bladder Adenocarcinoma and Ovarian Mucinous Adenocarcinoma.

CEACAM6 Crohn's Disease and Colorectal Cancer

CEL Maturity-Onset Diabetes Of The Young, Type 8, With Exocrine Dysfunction and Maturity-Onset Diabetes Of The Young.

CELF4 Benign Epilepsy With Centrotemporal Spikes.

CELSR3 Bladder Exstrophy-Epispadias-Cloacal Exstrophy Complex and Exstrophy Of Bladder.

CENPA Systemic Scleroderma and Rheumatic Disease.

CERS1 Epilepsy, Progressive Myoclonic, 8 and Myoclonus Epilepsy.

CFH Complement Factor H Deficiency and Hemolytic Uremic Syndrome, Atypical 1.

CFTR Cystic Fibrosis and Vas Deferens, Congenital Bilateral Aplasia Of.

CHD5 Neuroblastoma.

CHKA Large Cell Carcinoma With Rhabdoid Phenotypeand Myositis Fibrosa.

CHL1 3P- Syndrome and Large Cell Carcinoma With Rhabdoid Phenotype.

CHP2 Hepatocellular Carcinoma.

CHRM2 Major Depressive Disorder and Intestinal Schistosomiasis CHRNA3 Smoking As A Quantitative Trait Locus 3 and Autosomal Dominant Nocturnal Frontal Lobe Epilepsy.

CHRNB2 Epilepsy, Nocturnal Frontal Lobe, 3 and Chmb2-Related Nocturnal Frontal Lobe Epilepsy, Autosomal Dominant.

CHRNB3 Duane Retraction Syndromeand Cocaine Dependence.

CHRNB4 Substance Dependence and Tobacco Addiction.

CHST3 Spondyloepiphyseal Dysplasia With Congenital Joint Dislocations and Multiple Joint Dislocations, Short Stature, And Craniofacial Dysmorphism With Or Without Congenital Heart Defects.

CIDEB Specific Language Impairment.

CILP Intervertebral Disc Disease and Osteoarthritis.

CKAP2L Filippi Syndrome and Chromosome 16P13.3 Deletion Syndrome, Proximal.

CKMT1B Prostate Rhabdomyosarcoma and Dressleris Syndrome.

CLDN1 Ichthyosis, Leukocyte Vacuoles, Alopecia, And Sclerosing Cholangitisand Sclerosing Cholangitis.

CLRN1 Usher Syndrome, Type 3A and Retinitis Pigmentosa 61.

CNIH2 Schizophrenia.

CNNM1 Urofadal Syndrome 1.

CNTFR Cold-Induced Sweating Syndrome and Attention Deficit-Hyperactivity Disorder.

CNTN2 Epilepsy, Familial Adult Myoclonic, 5 and Benign Adult Familial Myoclonic Epilepsy.

CNTN4 Spinocerebellar Ataxia Type 16 and Chromosome 3Pter-P25 Deletion Syndrome.

CNTN6 Autonomic Nervous System Neoplasm and Peripheral Nervous System Neoplasm.

CNTNAP2 Pitt-Hopkins-Like Syndrome 1

CNTNAP3B Exstrophy Of Bladder.

CNTNAP4 Posterior Cortical Atrophy and Mowat-Wilson Syndrome.

CNTNAP5 Posterior Cortical Atrophy and Mowat-Wilson Syndrome.

COMT Schizophrenia and Panic Disorder 1.

C0R01A Immunodeficiency 8 and Coronin-1A Deficiency.

CPLX2 Huntington Disease and Schizophrenia.

CPLX3 Chromosome 15Q24 Deletion Syndrome. CPT1B Carnitine Palmitoyltransferase I Deficiencyand Visceral Steatosis.

CR2 Immunodeficiency, Common Variable, 7 and Systemic Lupus Erythematosus 9.

CRABP2 Embryonal Carcinoma and Basal Cell Carcinoma.

CRB1 Retinitis Pigmentosa 12 and Leber Congenital Amaurosis 8.

CRB2 Ventriculomegaly With Cystic Kidney Disease and Focal Segmental Glomerulosclerosis 9.

CREB3L3 Hyperlipoproteinemia, Type V and Hepatocellular Carcinoma.

CRTAC1 Bone Fracture.

CRX Cone-Rod Dystrophy 2 and Leber Congenital Amaurosis 7.

CSF1 Pigmented Villonodular Synovitis and Tenosynovial Giant Cell Tumor.

CSF1R Leukoencephalopathy, Hereditary Diffuse, With Spheroids and Anaplastic Large Cell Lymphoma.

CSF3R Neutropenia, Severe Congenital, 7, Autosomal Recessive and Neutrophilia, Hereditary.

CSMD2 Benign Adult Familial Myoclonic Epilepsyand Long Qt Syndrome 1.

CSMD3 Benign Adult Familial Myoclonic Epilepsyand Trichorhinophalangeal Syndrome, Type li.

CSPG5 Spontaneous Ocular Nystagmus and Kabuki Syndrome 1.

CTSK Pycnodysostosis and Endosteal Hyperostosis, Autosomal Dominant.

CTSS Cercarial Dermatitis and Mandibular Cancer.

CXADR Endotheliitis and Paracoccidioidomycosis.

CXCL13 Angioimmunoblastic T-Cell Lymphomaand Burkitt Lymphoma.

CXCL16 Angioimmunoblastic T-Cell Lymphomaand Burkitt Lymphoma.

CYP1B1 Glaucoma 3, Primary Congenital, A and Anterior Segment Dysgenesis 6.

CYP26B1 Radiohumeral Fusions With Other Skeletal And Craniofacial Anomalies and Occipital Encephalocele.

DBC1 Bladder Cancer and Transitional Cell Carcinoma.

DCX Lissencephaly, X-Linked, 1 and Subcortical Band Heterotopia.

DDC Aromatic L-Amino Acid Decarboxylase Deficiency and Oculogyric Crisis.

DDX3Y Spenmatogenic Failure, Y-Linked, 2 and Male Infertility.

DEFB1 Endophthalmitis and Tonsillitis.

DES Myopathy, Myofibrillar, 1 and Scapuloperoneal Syndrome, Neurogenic, Kaeser Type DGCR6 Velocardiofacial Syndrome and Digeorge Syndrome.

DGKH Adrenal Medulla Cancer and Extra-Adrenal Pheochromocytoma.

DI02 Graves' Disease and Euthyroid Sick Syndrome.

DISC1 Schizophrenia 9 and Schizophrenia.

DLG3 X-Linked Non-Specific Intellectual Disability and Non-Syndromic Intellectual Disability.

DLL4 Adams-Oliver Syndrome 6 and Adams-Oliver Syndrome.

DMGDH Dimethylglycine Dehydrogenase Deficiencyand Sarcosinemia.

DMXL2 Polyendocrine-Polyneuropathy Syndrome and Deafness, Autosomal Dominant 71

DNAH11 Ciliary Dyskinesia, Primary, 7 and Primary Ciliary Dyskinesia.

DNAH6 Primary Ciliary Dyskinesia.

DNAH9 Cardiac Tamponade and Primary Ciliary Dyskinesia.

DNAI1 Ciliary Dyskinesia, Primary, 1 and Kartagener Syndrome.

DNASE1L1 Human Monocytic Ehrlichiosis and Xerophthalmia.

DNM3 Optic Atrophy 1.

DPF1 Gastric cancer.

DPYD Dihydropyrimidine Dehydrogenase Deficiencyand Herpes Zoster.

DPYSL2 Dihydropyrimidine Dehydrogenase Deficiencyand Herpes Zoster.

DRD5 Blepharospasm, Benign Essential and Blepharospasm.

DSC2 Arrhythmogenic Right Ventricular Dysplasia, Familial, 11 and Familial Isolated Arrhythmogenic Ventricular Dysplasia, Right Dominant Form.

DSG2 Arrhythmogenic Right Ventricular Dysplasia, Familial, 10 and Cardiomyopathy, Dilated, 1Bb.

DSPP Dentinogenesis Imperfecta, Shields Type lii and Dentin Dysplasia, Type li.

DUSP4 Amyotrophic Lateral Sclerosis 11 and Echolalia.

DYDC2 Arrhythmogenic Right Ventricular Cardiomyopathy.

EBI3 Inflammatory Bowel Disease.

EDN1 Question Mark Ears, Isolated and Auriculocondylar Syndrome 3.

EEF1A2 Epileptic Encephalopathy, Early Infantile, 33 and Mental Retardation, Autosomal Dominant 38

EFHC2 Leukocoria and Turner Syndrome. EGF Hypomagnesemia 4, Renal and Familial Primary Hypomagnesemia With Normocalciuria And Normocalcemia.

EHBP1 Prostate Cancer, Hereditary, 12 and Prostate Cancer.

EMP1 Endobronchial Lipoma.

EMX2 Schizencephaly and Acquired Schizencephaly.

ENC1 Neuroblastoma.

ENG Telangiectasia, Hereditary Hemorrhagic, Type 1 and Hereditary Hemorrhagic Telangiectasia.

ENKUR Visceral Heterotaxy.

EN02 Granular Cell Tumor and Neuroendocrine Tumor.

ENPP7 Colorectal Cancer.

ENTPD1 Spastic Paraplegia 64, Autosomal Recessive and Proctitis.

ENTPD2 Dentin Sensitivity.

EPB41L4A Mixed Germ Cell Cancer.

EPB49 Hypotrichosis and Hereditary Spherocytosis.

EPDR1 Colorectal Cancer and Long Qt Syndrome 1.

EPHA6 Oculoauricular Syndrome.

EPHB2 Prostate Cancer/Brain Cancer Susceptibility and Prostate Cancer.

EPS8 Deafness, Autosomal Recessive 102 and Autosomal Recessive Non- Syndromic Sensorineural Deafness Type Dfnb

EPSTI1 Lupus Erythematosus and Systemic Lupus Erythematosus.

EVC2 Ellis-Van Creveld Syndrome and Weyers Acrofacial Dysostosis.

EYA4 Cardiomyopathy, Dilated, 1J and Deafness, Autosomal Dominant 10.

F10 Factor X Deficiency and Hemorrhagic Disease.

F7 Factor Vii Deficiency and Myocardial Infarction.

FAM107A Neuroblastoma and Brain Cancer.

FAM126A Leukodystrophy, Hypomyelinating, 5 and Hypomyelinating Leukodystrophy.

FAM155B Marantic Endocarditis and Enterobiasis.

FAM163A Neuroblastoma.

FAM5C Tongue Squamous Cell Carcinoma and Myocardial Infarction.

FAM64A Suppurative Periapical Periodontitisand Clonorchiasis.

FAM83D Kleine-Levin Hibernation Syndrome. FANCB Fanconi Anemia, Complementation Group B and Vacterl With Hydrocephalus.

FERMT3 Leukocyte Adhesion Deficiency

FFAR2 Lissencephaly 1 and Schizophrenia.

FGF12 Epileptic Encephalopathy, Early Infantile, 47 and Undetermined Early- Onset Epileptic Encephalopathy

FGF13 X-Linked Congenital Generalized Hypertrichosis and Wildervanck Syndrome.

FGF17 Hypogonadotropic Hypogonadism 20 With Or Without Anosmia and Normosmic Congenital Hypogonadotropic Hypogonadism.

FGFR3 Achondroplasia and Hypochondroplasia.

FLVCR1 Ataxia, Posterior Column, With Retinitis Pigmentosa and Posterior Column Ataxia.

FSHR Ovarian Hyperstimulation Syndrome and Ovarian Dysgenesis 1.

FSIP2 Spermatogenic Failure 34.

FUCA1 Fucosidosis and Lysosomal Storage Disease.

FUT9 Placental Malaria Infection

FXYD5 Leukemia, Acute Myeloid.

GAB1 Deafness, Autosomal Recessive 26 and Leopard Syndrome.

GABBR2 Epileptic Encephalopathy, Early Infantile, 59 and Neurodevelopmental Disorder With Poor Language And Loss Of Hand Skills.

GABRA5 Angelman Syndrome and Childhood Absence Epilepsy.

GAD1 Cerebral Palsy, Spastic Quadriplegic, 1 and Inherited Congenital Spastic Tetraplegia.

GAD2 Stiff-Person Syndrome and Autoimmune Polyendocrine Syndrome, Type li.

GAL3ST4 Pectus Excavatum.

GAP43 Developmental Coordination Disorder and Myositis Fibrosa.

GAR1 Dyskeratosis Congenita and Spinal Muscular Atrophy.

GASS Autoimmune Disease and Malignant Pleural Mesothelioma.

GATM Cerebral Creatine Deficiency Syndrome 3 and Astrocytoma.

GCNT1 Mast Cell Neoplasm.

GDAP1 Charcot-Marie-Tooth Disease, Type 4A and Charcot-Marie-Tooth Disease, Recessive Intermediate A. GDF5 Brachydactyly, Type A2 and Brachydactyly, Type C.

GEMIN4 Neurodevelopmental Disorder With Microcephaly, Cataracts, And Renal Abnormalities and Microcephaly.

GJA1 Oculodentodigital Dysplasia and Syndactyly, Type lii.

GLT1D1 Hepatocellular Carcinoma.

GLT8D2 Glycosyltransferase 8 Domain Containing 2

GLYATL2 Mitochondrial acyltransferase which transfers the acyl group to the N- terminus of glycine. Conjugates numerous substrates, such as arachidonoyl-CoA and saturated medium and long-chain acyl-CoAs ranging from chain-length C8:0-CoA to C18:0-CoA, to form a variety of N- acylglycines.

GNA14 Kaposiform Hemangioendothelioma and Angioma, Tufted.

GPD1 Hypertriglyceridemia, Transient Infantileand Brugada Syndrome.

GPI Hemolytic Anemia, Nonspherocytic, Due To Glucose Phosphate Isomerase Deficiency and Glucose Phosphate Isomerase Deficiency.

GPR64 Vas Deferens, Congenital Bilateral Aplasia Of, X-Linked and Vas Deferens, Congenital Bilateral Aplasia Of.

GPR98 Usher Syndrome, Type lie and Febrile Seizures, Familial, 4.

GPX4 Spondylometaphyseal Dysplasia, Sedaghatian Type and Neurotic Disorder.

GRIA1 Status Epilepticus and Fragile X Syndrome.

GRIA2 Status Epilepticus and Lateral Sclerosis.

GRIA3 Mental Retardation, X-Linked, Syndromic, WU Type and Rasmussen Encephalitis.

GRIK3 Schizophrenia.

GRIN2B Mental Retardation, Autosomal Dominant 6, With Or Without Seizures and Epileptic Encephalopathy, Early Infantile, 27.

GRM1 Spinocerebellar Ataxia, Autosomal Recessive 13 and Spinocerebellar Ataxia 44.

GRM4 Epilepsy, Idiopathic Generalized 10 and Schizophrenia.

GRM7 Age-Related Hearing Loss and Lubs X-Linked Mental Retardation Syndrome.

GRPR Agoraphobia and Suppression Of Tumorigenicity 12.

GSC Short Stature, Auditory Canal Atresia, Mandibular Hypoplasia, And Skeletal Abnormalities and Synostosis. GSTA1 Ovarian Endodermal Sinus Tumor and Ovarian Primitive Germ Cell Tumor

GSTM1 Senile Cataract and Asbestosis.

GSTO2 Parkinson Disease, Late-Onset.

GSTT1 Larynx Cancer and Senile Cataract.

GSTT2 Colon Adenoma and Deafness, Autosomal Recessive 12.

GUCY2C Meconium Ileus and Diarrhea 6.

GUCY2D Cone-Rod Dystrophy 6 and Leber Congenital Amaurosis 1

GYLTL1B Interstitial Myocarditis and Muscular Dystrophy-Dystroglycanopathy , Type

B, 6.

H19 Wilms Tumor 2 and Beckwith-Wiedemann Syndrome.

HAVCR2 Hepatitis A and Hepatitis.

HERC6 Meningococcal Meningitis.

HESX1 Septooptic Dysplasia and Pituitary Stalk Interruption Syndrome.

HIP1R Cataract 8, Multiple Types and Parkinson Disease, Late-Onset.

HIST1H3C Diffuse Intrinsic Pontine Glioma.

HIVEP2 Mental Retardation, Autosomal Dominant 43 and Hivep2-Related Intellectual Disability.

HK1 Hemolytic Anemia, Nonspherocytic, Due To Hexokinase

Deficiency and Neuropathy, Hereditary Motor And Sensory, Russe Type.

HK2 Pediatric Osteosarcoma and Chondroblastoma.

HLA-A Sarcoidosis 1 and Multiple Sclerosis

HLA-C Psoriasis 1 and Human Immunodeficiency Virus Type 1.

HLA-DRA Graham-Little-Piccandi-Lassueur Syndrome and Heart Lymphoma.

HMGCR Hyperlipidemia, Familial Combined and Marek Disease.

HNF1B Renal Cysts And Diabetes Syndrome and Diabetes Mellitus, Noninsulin- Dependent.

HNMT Mental Retardation, Autosomal Recessive 51 and Asthma.

HOMER1 Ogden Syndrome.

HPCAL4 Holoprosencephaly 3.

HPD Tyrosinemia, Type lii and Hawkinsinuria.

HPGD Digital Clubbing, Isolated Congenitaland Hypertrophic Osteoarthropathy, Primary, Autosomal Recessive, 1. HSPG2 Schwartz-Jampel Syndrome, Type 1 and Dyssegmental Dysplasia, Silverman-Handmaker Type.

HTR2A Major Depressive Disorder and Obsessive-Compulsive Disorder.

HTR2C Anxiety and Premature Ejaculation.

ID01 Listeriosis and Bladder Disease.

IFI16 Neonatal Adrenoleukodystrophy.

IFI30 Atrophic Rhinitis.

IFIT3 Systemic Lupus Erythematosus.

IFLTD1 Respiratory System Benign Neoplasm.

IFNA1 Hepatitis C and Hepatitis.

IFNA17 Adenosquamous Pancreas Carcinoma and Crimean-Congo Hemorrhagic Fever.

IGF1 Insulin-Like Growth Factor I and Pituitary Gland Disease.

IGFBP2 Malignant Ovarian Cyst and Insulin-Like Growth Factor I.

IHH Acrocapitofemoral Dysplasia and Brachydactyly, Type A1.

IL1B Gastric Cancer, Hereditary Diffuse and Periodontal Disease.

IL1R1 Schnitzler Syndrome and Cinca Syndrome.

IL1RAPL1 Mental Retardation, X-Linked 21 and X-Linked Non-Specific Intellectual Disability.

IL1RAPL2 Cold Urticaria and Anterior Scleritis.

IL26 Inflammatory Bowel Disease.

IL2RB Oligoarticular Juvenile Idiopathic Arthritis and Rheumatoid Factor- Negative Juvenile Idiopathic Arthritis.

IL34 Chronic Apical Periodontitis

IL6R Castleman Disease and Pycnodysostosis.

IMPG2 Macular Dystrophy, Vitelliform, 5 and Retinitis Pigmentosa 56

INA Wernicke Encephalopathy and Medulloepithelioma.

INHBA Ovary Adenocarcinoma and Preterm Premature Rupture Of The Membranes

INPP4B Vulva Adenocarcinoma.

INSM2 Insulinoma.

IRF5 Systemic Lupus Erythematosus 10 and Inflammatory Bowel Disease 14.

IRF6 Popliteal Pterygium Syndrome and Van Der Woude Syndrome 1. IRF8 Immunodeficiency 32A and Immunodeficiency 32B.

IRX5 Hamamy Syndrome and Griscelli Syndrome, Type 3.

ITGA11 Tick Infestation and Parasitic Ectoparasitic Infectious Disease.

ITGA2 Bleeding Disorder, Platelet-Type, 9 and Fetal And Neonatal Alloimmune Thrombocytopenia.

ITGA8 Renal Hypodysplasia/Aplasia 1 and Renal Agenesis, Bilateral.

ITGB8 Arteriovenous Malformation.

IYD Thyroid Dyshormonogenesis 4 and Familial Thyroid Dyshormonogenesis.

JAG1 Alagille Syndrome 1 and Tetralogy Of Fallot.

JMJD6 Deep Angioma and Intramuscular Hemangioma.

KAZALD1 Lobar Holoprosencephalyand Pleural Cancer.

KBTBD8 Treacher Collins Syndrome 1.

KCNA4 Episodic Ataxia, Type 1 and Episodic Ataxia.

KCND2 Cycloplegia and Gastrointestinal Lymphoma.

KCNF1 Deafness, Autosomal Recessive 47.

KCNH3 Background Diabetic Retinopathy.

KCNH6 Charcot-Marie-Tooth Disease, Demyelinating, Type 1D and Charcot- Marie-Tooth Disease, Axonal, Type 2F.

KCNIP2 Spinocerebellar Ataxia Type 19/22 and Brugada Syndrome.

KCNJ13 Snowflake Vitreoretinal Degeneration and Leber Congenital Amaurosis

16.

KCNJ2 Andersen Cardiodysrhythmic Periodic Paralysis and Short Qt Syndrome

3.

KCNMA1 Amyloid Accumulation Drives Proteome-wide Alterations in Mouse Models of Alzheimer’s Diseaselike Pathology

KCNN3 Retinitis Pigmentosa 19 and Spinocerebellar Ataxia 2.

KCTD12 Gastrointestinal Stromal Tumor.

KCTD13 Schizophreniaand Psychotic Disorder.

KIAA0226L Cervical Cancer.

KIAA0319 Dyslexia 2 and Dyslexia.

KIAA1324 Uterine Corpus Serous Adenocarcinoma and Estrogen Excess.

KIFAP3 Progressive Bulbar Palsy and Amyotrophic Lateral Sclerosis 1.

KLF10 Hemoglobinopathy and Pancreatic Cancer. KLHL1 Spinocerebellar Ataxia 8.

KLHL7 Retinitis Pigmentosa 42 and Cold-Induced Sweating Syndrome 3.

KLK6 Colon Adenoma and Synucleinopathy.

KPNA2 Malignant Germ Cell Tumor and Ovarian Endodermal Sinus Tumor.

KRT18 Cryptogenic Cirrhosis and Epithelioid Trophoblastic Tumor.

KRT23 Colonic Benign Neoplasm.

KRT7 Cystadenoma and Adenosquamous Carcinoma.

LAMA2 Muscular Dystrophy, Congenital Merosin-Deficient, 1Aand Congenital Muscular Dystrophy Type 1 A.

LAMA4 Cardiomyopathy, Dilated, 1 Jj and Familial Isolated Dilated Cardiomyopathy.

LAPTM5 Charcot-Marie-Tooth Disease, Dominant Intermediate C and Charcot- Marie-Tooth Disease Intermediate Type.

LATS2 Intracranial Abscess.

LCE4A Precursors of the comified envelope of the stratum comeum.

LCN9 Parasitic Ectoparasitic Infectious Disease.

LMAN1 Factor V And Factor Viii, Combined Deficiency Of, land Factor V And Factor Viii, Combined Deficiency Of, 2.

LM01 Exencephaly and T-Cell Leukemia.

LM07 Townes-Brocks Syndrome.

LPL Hyperlipoproteinemia, Type I and Hyperlipidemia, Familial Combined.

LRAT Leber Congenital Amaurosis 14 and Severe Eady-Childhood-Onset Retinal Dystrophy.

LRRC10 Dilated Cardiomyopathy.

LRRC48 Primary Ciliary Dyskinesia.

LRRC7 Dental Pulp Necrosis and Dental Pulp Disease.

LYPD6B Tobacco Addiction.

MAGEA5 Melanoma and Dyskeratosis Congenita.

MAGI2 Nephrotic Syndrome 15 and Chromosome 1P36 Deletion Syndrome.

MAMLD1 Hypospadias 2, X-Linked and Hypospadias.

MAOB Nome Disease and Postencephalitic Parkinson Disease.

MAP1LC3A Leber Congenital Amaurosis 6 and Lacrimal Gland Adenocarcinoma.

MAPK8 Fatty Liver Disease and Renal Fibrosis. MAPK8IP1 Diabetes Mellitus, Noninsulin-Dependent and Sarcomatoid Squamous Cell Skin Carcinoma.

MEGF10 Myopathy, Areflexia, Respiratory Distress, And Dysphagia, Early-Onset and Dysphagia.assodated with schizophrenia, Areflexia, Respiratory Distress, And Dysphagia, Early-Onset and Dysphagia

MLC1 Megalencephalic Leukoencephalopathy With Subcortical Cysts and Mld- Related Megalencephalic Leukoencephalopathy With Subcortical Cysts.

MMP13 Spondyloepimetaphyseal Dysplasia, Missouri Typeand Metaphyseal Dysplasia, Spahr Type.

MTS Alzheimer Disease and Amyotrophic Lateral Sclerosis 1.

MTTP Abetalipoproteinemia and Abdominal Obesity-Metabolic Syndrome 1.

MX1 Influenza and Viral Encephalitis.

NEFM Pineal Parenchymal Tumor Of Intermediate Differentiationand Wallerian Degeneration.

NOS2 Malaria and Meningioma, Radiation-Induced.

NPAS3 Holoprosencephaly 8 and Schizophrenia.

NPHP1 Senior-Loken Syndrome 1 and Nephronophthisis 1.

NPNT Fraser Syndrome 1.

NPPC Achondroplasia and Paraphimosis.

NR1H3 Multiple Sclerosis and Cerebrotendinous Xanthomatosis.

NR1I2 Cerebrotendinous Xanthomatosis and Biliary Tract Disease.

NR2E1 Lipodystrophy, Familial Partial, Type 3.

NR4A2 Parkinson Disease, Late-Onset and Chondrosarcoma, Extraskeletal Myxoid.

NRG1 Schizophrenia and Schizophreniform Disorder.

NTFS Hypochondriasis and Diabetic Polyneuropathy.

NTS Duodenogastric Reflux and Dumping Syndrome.

OAS3 Chikungunya and Tick-Borne Encephalitis.

OAT Gyrate Atrophy Of Choroid And Retina and Choroid Disease.

TENM1 Anosmia, Isolated Congenital and Anal Margin Carcinoma.

TENM3 Microphthalmia, Isolated, With Coloboma 9and Colobomatous Microphthalmia.

OSCP1 Nasopharyngeal Carcinoma and Pharynx Cancer. 07X2 Microphthalmia, Syndromic 5 and Pituitary Hormone Deficiency, Combined, 6.

P2RY12 Bleeding Disorder, Platelet-Type, 8 and Drug Metabolism, Poor, Cyp2c19- Related

PAH Phenylketonuria and Mild Phenylketonuria.

PAM Phaeohyphomycosis and Menkes Disease.

PAPSS2 Brachyolmia Type 4 With Mild Epiphyseal And Metaphyseal Changes and Brachyolmia.

PCDH11X Dyslexia and Schizoaffective Disorder.

PCDH18 Hemophagocytic Lymphohistiocytosis and Patent Foramen Ovale.

PCGF5 Interleukin-7 Receptor Alpha Deficiency.

PCNT Microcephalic Osteodysplastic Primordial Dwarfism, Type li and Seckel Syndrome.

PCSK9 Hypercholesterolemia, Autosomal Dominant, 3 and Homozygous Familial Hypercholesterolemia.

PDCD6IP Adult Neuronal Ceroid Lipofuscinosis

PDE5A Priapism and Nonarteritic Anterior Ischemic Optic Neuropathy.

PDGFRL Colorectal Cancer and Hepatocellular Carcinoma.

PDIA2 Alpha Thalassemia-Intellectual Disability Syndrome Type 1 and Multiple Sulfetase Deficiency.

PGAM1 Phosphoglycerate Mutase Deficiency

PHOX2B Central Hypoventilation Syndrome, Congenitaland Neuroblastoma 2.

PIS Myeloid Tumor Suppressor and Chorioangioma.

PIEZ01 Dehydrated Hereditary Stomatocytosis 1 With Or Without Pseudohyperkalemia And/Or Perinatal Edema and Lymphedema, Hereditary, lii.

PIEZ02 Marden-Walker Syndrome and Arthrogryposis, Distal, Type 3.

PIPOX Peroxisomal Biogenesis Disorders.

PLA2G1B Distal Hereditary Motor Neuropathy, Type liand Neurodegeneration With Brain Iron Accumulation 2B.

PLA2G7 Platelet-Activating Factor Acetylhydrolase Deficiency and Atopy.

PLB1 PLB1 include Amyotrophic Lateral Sclerosis 3 and Opportunistic Mycosis.

PLCG2 Autoinflammation, Antibody Deficiency, And Immune Dysregulation, Plcg2-Associated and Familial Cold Autoinfiammatory Syndrome 3 PLLP Bardet-Biedl Syndrome

PLP1 Pelizaeus-Merzbacher Disease and Spastic Paraplegia 2, X- Linked. include Spastic Paraplegia 2, X-Linked and Pelizaeus-Merzbacher Disease, myelin sheaths, as well as in oligodendrocyte development and axonal survival

PLXNA4 Cerebral Amyloid Angiopathy, Itm2b-Related, 1.

PNCK Salivary Gland Carcinomaand Salivary Gland Disease.

PNOC Pain Agnosia and Agnosia.

PODXL Atypical Juvenile Parkinsonism and Parkinson Disease 2, Autosomal Recessive Juvenile.

POU2F2 Papilloma and T-Cell/Histiocyte Rich Large B Cell Lymphoma.

POU3F3 Esophageal Cancer and Central Nervous System Tuberculosis

PPARD Diabetic Cataract and Abdominal Obesity-Metabolic Syndrome Quantitative Trait Locus 2.

PPARGC1A Obesity and Lipomatosis.

PRDM16 acute myeloid leukemia

PRKCB Papillary Glioneuronal Tumors and Choidoid Glioma.

PRKG2 Chromosome 4Q21 Deletion Syndromeand Malignant Hemangioma.

PRL Pituitary Gland Disease and Empty Sella Syndrome.

PRODH Hyperprolinemia, Type I and Schizophrenia 4.

PRRX1 Agnathia-Otocephaly Complex and Dysgnathia Complex.

PSD Immunodeficiency 10 and Branch Retinal Artery Occlusion.

PTCHD1 Autism X-Linked 4 and Autism Spectrum Disorder.

PTGER2 Asthma, Nasal Polyps, And Aspirin Intoleranceand Deafness, Autosomal Dominant 17.

PTGIR Erythroleukemia, Familial and Cone-Rod Dystrophy 10.

PTGS2 Stomach Disease and Peptic Ulcer Disease.

PTK2B Cone-Rod Dystrophy 5 and Transient Cerebral Ischemia.

PTN Noma

PTPRQ Deafness, Autosomal Recessive 84A and Deafness, Autosomal Dominant

73.

PTPRR Deafness, Autosomal Recessive 84A.

PTPRZ1 Perrault Syndrome 1 and Hyperlysinemia, Type I.

PVALB Fish Allergy and Fetal Alcohol Syndrome. RAB3A Cone-Rod Dystrophy 7 and Isolated Growth Hormone Deficiency, Type II.

RAPGEF4 Lesch-Nyhan Syndrome and Noonan Syndrome 1.

RASIP1 Enamel Erosion and Tooth Erosion.

RASL12 Nemaline Myopathy 6 and Zika Fever.

RBMXL2 Cardiomyopathy, Dilated, 3B.

RBP3 Retinitis Pigmentosa 66 and Rbp3-Related Retinitis Pigmentosa.

RDH5 Fundus Albipunctatus and Rdh5-Related Fundus Albipunctatus.

REEP1 Spastic Paraplegia 31 , Autosomal Dominant and Neuronopathy, Distal Hereditary Motor, Type Vb.a Neurodegenerative Disorder.

REG3A Pancreatitis and Acute Pancreatitis.

RGS6 Hirschsprung Disease 1 and Alcoholic Cardiomyopathy.

RGS7 Retinoschisis 1 , X-Linked, Juvenile.

RLTPR Immunodeficiency 58and Combined Immunodeficiency, X-Linked.

RNASE2 Peripheral Demyelinating Neuropathy, Central Dysmyelination, Waardenburg Syndrome, And Hirschsprung Disease and Lacrimoauriculodentodigital Syndrome

RNF212 Recombination Rate Quantitative Trait Locus 1.

ROBG3 Gaze Palsy, Familial Horizontal, With Progressive Scoliosis, 1 and Horizontal Gaze Palsy With Progressive Scoliosis.

RPE65 Retinitis Pigmentosa 20 and Leber Congenital Amaurosis 2.

RPH3AL Medulloblastoma.

RTN4R Schizophrenia and Acute Lymphocytic Leukemia.

RUNX3 Testicular Yolk Sac Tumor and Esophagus Squamous Cell Carcinoma.

RWDD2B Monosomy 21.

S100A14 Small Intestine Adenocarcinoma.

SATB2 Glass Syndrome and Cleft Palate, Isolated.

SCARF1 Syndromic X-Linked Intellectual Disability Snyder Type and Urethral Stricture.

SCD5 Chromosome 4Q21 Deletion Syndrome and Lipodystrophy, Congenital Generalized, Type 3.

SCN1B Epileptic Encephalopathy, Early Infantile, 52 and Generalized Epilepsy With Febrile Seizures Plus, Type 1

SCN2A Seizures, Benign Familial Infantile, 3 and Epileptic Encephalopathy, Early Infantile, 11. SCN2B Atrial Fibrillation, Familial, 14and Familial Atrial Fibrillation.

SDPR Well-Differentiated Liposarcoma.

SECTM1 Arthus Reaction and Cryptococcosis.

SELL Arthus Reaction and Cryptococcosis.

SFRP4 Pyle Disease and Osteomalacia.

SH3BP2 Cherubism and Giant Cell Reparative Granuloma.

SH3KBP1 Breast Adenocarcinomaand Adrenal Cortical Adenocarcinoma.

SH3TC1 Neuropathy, Congenital Hypomyelinating Or Amyelinating, Autosomal Recessive.

SHANK1 Autism Spectrum Disorder and Diabetic Encephalopathy.

SHROOM2 Ocular Albinism.

SIM2 Down Syndrome and Holoprosencephaly 1.

SLC12A5 Solute Carrier Family 12 Member 5

SLC16A10 Thyroid hormone signaling pathway

SLC16A14 Proton-linked monocarboxylate transporter.

SLC17A6 Gnathodiaphyseal Dysplasia and Tendinosis.

SLC1A2 Epileptic Encephalopathy, Early Infantile, 41 and Wernicke Encephalopathy

SLC1A3 Episodic Ataxia, Type 6 and Episodic Ataxia.

SLC24A2 Brain Injury and Achromatopsia.

SLC26A2 Achondrogenesis, Type lb and Epiphyseal Dysplasia, Multiple, 4.

SLC2A4 Diabetes Mellitus, Noninsulin-Dependentand Diabetes Mellitus.

SLC34A2 Pulmonary Alveolar Microlithiasis and Testicular Microlithiasis.

SLC41A1 Nephronophthisis.

SLC4A5 Alstrom Syndrome.

SLC6A1 Myoclonio-Atonic Epilepsy and Myoclonic-Astastic Epilepsy.

SLC6A15 Major Depressive Disorder.

SLC6A3 Parkinsonism-Dystonia, Infantile and Nicotine Dependence, Protection Against.

SLC7A14 Retinitis Pigmentosa 68 and Slc7a14-Related Retinitis Pigmentosa

SLC9A9 Autism 16 and Attention Deficit-Hyperactivity Disorder.

SLCC2B1 Persistent Fetal Circulation Syndrome. SLC04A1 Mucinous Cystadenocarcinoma and Aneurysmal Bone Cysts

SLC04C1 Eastern Equine Encephalitis.

SLCOSA1 Mesomelia-Synostoses Syndrome and Mesomelia.

SLIT1 Diaphragm Disease and Diaphragmatic Hernia, Congenital.

SMEK3P Protein Phosphatase 4 Regulatory Subunit 3C

SNAP25 Myasthenic Syndrome, Congenital, 18and Presynaptic Congenital Myasthenic Syndromes.

SNTG1 Idiopathic Scoliosis and Basal Ganglia Calcification.

SORCS1 Narcolepsy.

SP100 Primary Biliary Cirrhosis and Autoimmune Disease Of Urogenital Tract.

SPAG5 Patellar Tendinitis.

SPAG6 Hydrocephalus.

SPI1 Inflammatory Diarrhea and Neutrophil-Specific Granule Deficiency.

SPTBN1 Beckwith-Wiedemann Syndrome.

SPTBN4 Myopathy, Congenital, With Neuropathy And Deafness and Myopathy, Congenital. Spectrin b-lll

SSTR1 Acromegaly and Pituitary Adenoma.

SSTR2 Pancreatic Endocrine Carcinoma and Type C Thymoma.

SSTR3 Pituitary Adenoma, Prolactin-Secreting and Oncogenic Osteomalacia.

ST8SIA2 Osteogenesis Imperfecta, Type Xv and Eumycotic Mycetoma.

STAB1 Bacillary Angiomatosis and Histiocytosis.

STMN2 Goldberg-Shprintzen Syndrome and Creutzfeldt-Jakob Disease.

STOML3 Gliosarcoma.

STXBP1 Epileptic Encephalopathy, Early Infantile, 4 and Epileptic Encephalopathy, Early Infantile, 15.

SULF1 Mesomelia-Synostoses Syndrome and Mesomelia.

SULT1E1 Anteroseptal Myocardial Infarctionand Inferior Myocardial Infarction.

SULT4A1 Anteroseptal Myocardial Infarction and Schizotypal Personality Disorder.

SYTL2 Angioedema and Griscelli Syndrome, Type 3.

TAC1 Bronchitis and Neurotrophic Keratopathy.

TACR3 Hypogonadotropic Hypogonadism 11 With Or Without

Anosmia and Normosmic Congenital Hypogonadotropic Hypogonadism.

TANK Vaccinia. TAS2R16 Alcohol Dependence and Alcohol Use Disorder.

TET2 Myelodysplastic Syndrome and Refractory Anemia.

TFF3 Colitis and Barrett Esophagus.

TGFBR2 Loeys-Dietz Syndrome 2 and Colorectal Cancer, Hereditary Nonpolyposis, Type 6.

THPO Thrombocythemia 1 and Essential Thrombocythemia.

THSD1 Intracranial Aneurysm and Cerebral Arterial Disease.

TMEM132D Rhirus Pubis Infestation and Lice Infestation.

TNFRSF9 Retroperitoneal Hemangiopericytoma and Colorectal Cancer.

TNFSF10 Malignant Glioma and Ulceroglandular Tularemia.

TNMD Age-related Macular Degeneration

TNN Adhesive Otitis Media and Chronic Purulent Otitis Media.

TNNT1 Nemaline Myopathy 5 and Nemaline Myopathy.

TPCN2 Skin/Hair/Eye Pigmentation, Variation In, 10 and Deafness, Autosomal Recessive 63.

TRAPPC3 Tietz Albinism-Deafness Syndrome and Cardiac Tamponade.

TREM2 Charcot-Ma rie-T ooth Disease, Axonal, Type 2R and Axonal Neuropathy.

TRIP13 Mosaic Variegated Aneuploidy Syndrome 3 and Mosaic Variegated Aneuploidy Syndrome.

TROAP Ectopic Pregnancy.

TRPM3 Dentin Sensitivity and Chronic Fatigue Syndrome.

TRPV3 Palmoplantar Keratoderma, Mutilating, With Periorificial Keratotic Plaques and Palmoplantar Keratoderma, Nonepidermolytic, Focal 2.

TSPAN13 Alzheimer's disease (cognitive decline) - Associated SNPs

TSPAN2 Focal demyelination associated with amyloid plaque formation in Alzheimer's disease Tetraspanin 2

TSPAN7 X-Linked Non-Specific Intellectual Disability and Acute Apical Periodontitis.

TSPO Hepatic Encephalopathy and Focal Epilepsy.

TTBK1 Childhood-Onset Schizophrenia and Alzheimer Disease.

TTC40 Cilia And Flagella Associated Protein 46

TTC8 Retinitis Pigmentosa 51 and Bardet-Biedl Syndrome 8.

TUBB2A Cortical Dysplasia, Complex, With Other Brain Malformations 5 and Tubulin, Beta. TYROBP Polycystic Lipomembranous Osteodysplasia With Sclerosing Leukoencephalopathy and Dementia.

UCHL1 Spastic Paraplegia 79, Autosomal Recessiveand Parkinson Disease 5, Autosomal Dominant.

UGT2B17 Bone Mineral Density Quantitative Trait Locus

12 and Osteoporosis .Alzheimer's disease and osteoporosis

UNC13A Amyotrophic Lateral Sclerosis 1 and Frontotemporal Dementia And/Or Amyotrophic Lateral Sclerosis 1.

UPK3B Signet Ring Cell Adenocarcinoma.

USP2 Ovarian Serous Cystadenocarcinoma and Serous Cystadenocarcinoma.

UTS2R Amyotrophic Lateral Sclerosis 3 and Pheochromocytoma.

VAMP2 Tetanus and Primary Bacterial Infectious Disease.

VASH2 Angiogenesis inhibitor.

VAV3 Glaucoma, Normal Tension.

VCAN Wagner Vitreoretinopathy and Wagner Syndrome.

VIL1 Type 1 Diabetes Mellitus 13 and Dacryoadenitis.

VLDLR Cerebellar Ataxia, Mental Retardation, And Dysequilibrium Syndrome 1 and Cerebellar Hypoplasia.

VPREB1 ConRJiobolomycosis and Mu Chain Disease.

VSNL1 Acute Encephalopathy With Biphasic Seizures And Late Reduced Diffusion and Alzheimer Disease.

WASF1 Wiskott-Aldrich Syndrome and Spinocerebellar Ataxia, Autosomal Recessive 1.

WDR16 Dextrocardia With Situs Inversus.

WDR63 Hemometra.

WDR91 Usher Syndrome, Type I.

WDR96 Spermatogenic Failure 19 and Non-Syndromic Male Infertility Due To Sperm Motility Disorder.

WIF1 Esophageal BasaloRJ Squamous Cell Carcinoma and Colorectal Cancer.

WNT10B Split-Hand/Foot Malformation 6 and Tooth Agenesis, Selective, 8.

WNT7A Fibular Aplasia Or Hypoplasia, Femoral Bowing And Poly-, Syn-, And Oligodactyly and Ulna And Fibula, Absence Of, With Severe Limb Deficiency.

WNT8B Gastric cancer.

WNT9A Gastric cancer. WT1 Wilms Tumor 1 and Denys-Drash Syndrome.

XIST X Inactivation, Familial Skewed, 1 and Hypogonadotropic Hypogonadism.

XKR4 X-Linked Kx Blood Group Related 4

XRRA1 X-Ray Radiation Resistance Associated 1

ZBTB16 Skeletal Defects, Genital Hypoplasia, And Mental Retardation and Acute Promyelocytic Leukemia

ZDBF2 Nasopalpebral Lipoma-Coloboma Syndromeand Coloboma Of Macula.

ZFHX3 Prostate Cancer and Atrial Fibrillation

MAK Retinitis Pigmentosa 62 and Mak-Related Retinitis Pigmentosa.

BID Cat eye syndrome

PIANP Diabetic Autonomic Neuropathy.

C2 Complement Component 2 Deficiency and Macular Degeneration, Age- Related, 14.

C0R07 Chromosomal Disease.

DGCR5 DiGeorge syndrome

GNG2 Hemiplegic Migraine.

TNFSF13 Brain Glioblastoma Multifbrme and Igg4-Related Disease.

TRPM1 Night Blindness, Congenital Stationary, Type 1C and Congenital Stationary Night Blindness.

KL Tumoral Calcinosis, Hyperphosphatemic, Familial, 3 and Tumoral Calcinosis, Hyperphosphatemic, Familial, 1.

IL10RB Rapidly Progressive Dementia as Presenting Feature in Inflammatory Bowel Disease; Inflammatory Bowel Disease 25, Early Onset, Autosomal Recessive

[00155] One of skill in the art will recognize that sequence data for the genes listed above can be obtained in publicly available gene databases such as GeneCards, GenBank, Malcard, Uniport and PathCard databases. The skilled worker will recognize these markers as set forth exemplarily herein to be human-specific marker proteins as identified, inter alia, in genetic information repositories such as GenBank; Accession Number for these markers are set forth in exemplary fashion in Table 7. One having skill in the art will recognize that variants derive from the full length gene sequence. Thus, the data findings and sequences in Table 7 encode the respective polypeptide having at least 70% homology to other variants, including full length sequences. Example 7: Neural Organoids for Testing Drug Efficacy.

[00156] Neural organoids can be used for pharmaceutical testing, safety, efficacy, and toxicity profiling studies. Specifically, using pharmaceuticals and human neural organoids, beneficial and detrimental genes and pathways associated with Alzheimer's disease can be elucidated. Neural organoids as provided herein can be used fortesting candidate pharmaceutical agents, as well as testing whether any particular pharmaceutical agent inter alia for Alzheimer's disease should be administered to a particular individual based on responsiveness, alternation, mutation, or changes in gene expression in a neural organoid produced from cells from that individual or in response to administration of a candidate pharmaceutical to said individual’s neural organoid.

Other Embodiments:

[00157] From the foregoing description, it will be apparent that variations and modifications can be made to the invention described herein to adopt it to various usages and conditions. Such embodiments are also within the scope of the following claims. [00158] The recitation of a listing of elements in any definition of a variable herein includes definitions of that variable as any single element or combination (or subcombination) of listed elements. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

[00159] All patents and publications mentioned in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference.

TABLE 8. SEQUENCE IDs for SEQUENCE LISTINGS RELATED TO ALZHEIMER’S DISEASE

SEQ ID NO: 49 BIN1

SEQ ID NO: 50 MEF2C

SEQ ID NO: 51 SCIMP

SEQ ID NO: 52 HLA-DRB5

SEQ ID NO: 53 HLA-DRB1

SEQ ID NO: 54 CD2AP

SEQ ID NO: 55 NME8

SEQ ID NO: 56 ZCWPW1

SEQ ID NO: 57 EPHA1 SEQ ID NO: 58 PTK2B

SEQ ID NO: 59 CLU

SEQ ID NO: 60 ECHDC3

SEQ ID NO: 61 PICALM

SEQ ID NO: 62 SORL1

SEQ ID NO: 63 FERMT2

SEQ ID NO: 64 SCL24A4

SEQ ID NO: 65 SPPL2A

SEQ ID NO: 66 HBEGF

SEQ ID NO: 67 CASS4

SEQ ID NO: 68 APOE

SEQ ID NO: 69 MS4A6A

SEQ ID NO: 70 HLA-DQA1

SEQ ID NO: 71 SQSTM1

SEQ ID NO: 72 UNC5C

SEQ ID NO: 73 AKAP9

SEQ ID NO: 74 PLD3

SEQ ID NO: 75 TRIP4

SEQ ID NO: 76 PLXNA4

SEQ ID NO: 77 MTHFR

SEQ ID NO: 78 TTC3

SEQ ID NO: 79 PSEN2

SEQ ID NO: 80 ZNF628

SEQ ID NO: 81 KCTD2

SEQ ID NO: 82 CYP2D6

SEQ ID NO: 83 ADAM 10

SEQ ID NO: 84 PSEN1

SEQ ID NO: 85 TREML2

SEQ ID NO: 86 ADNPP

SEQ ID NO: 87 POGZ

[00160] Having described the invention in detail and by reference to specific aspects and/or embodiments thereof, it will be apparent that modifications and variations are possible without departing from the scope of the invention defined in the appended claims. More specifically, although some aspects of the present invention may be identified herein as particularly advantageous, it is contemplated that the present invention is not limited to these particular aspects of the invention. Percentages disclosed herein can vary in amount by ±10, 20, or 30% from values disclosed and remain within the scope of the contemplated invention

Appendix: Brain Structure Markers and Accession No.

Gene Accession Gene Accession

Cerebellar Spinal Cord

ATOH1 , NM 005172.1 HOXA1 NM 005522.4

PAX6 NM 000280.4 HOXA2 NM 006735.3

SOX2 NM 003106.3 HOXA3 NM 030661.4

LHX2 NM 004789.3 HOXB4 NM 024015.4

GRID2 NM 001510.3 HOXAS NM 019102.3

HOSCS NM 018953.3

Dopaminergic HOXDI3 NM 000523.3

VMAT2 NM 003054.4

DAT NM 001044.4 GABAergic

D2 NM 000795.3 NKCCI NM 000338.2

KCC2 NM 001134771.1

Cortical

NeuN NM 001082575.2 Microglia

FOXP2 NM 014491.3 AIF1 NM 032955.2

CNTN4 NM 175607.2 CD4 NM 000616.4

TBR1 NM 004612.3

Brain Stem

Retinal FGF8 NM 033165.3

GUY2D NM 000180.3 INSM1 NM 002196.2

GUY2F NM 001522.2 GATA2 NM 001145661.1

RAX NM 013435.2 ASCL1 NM 004316.3

GATA3 NM 001002295.1

Granular Neuron

SOX2 NM 003106.3

NeuroDI NM 002500.4

DCX NM 000555.3

EMX2 NM 000555.3 FOXG1 NM 005249.4

PROX1 NM 001270616.1