Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR TREATING AMYOTROPHIC LATERAL SCLEROSIS
Document Type and Number:
WIPO Patent Application WO/2015/051283
Kind Code:
A1
Abstract:
Methods for modulating expression of SMN1 and/or SMN2 in cells obtained from subjects having ALS or in subjects having ALS using single stranded oligonucleotides are provided. Methods for treating ALS using single stranded oligonucleotides are also provided.

Inventors:
KRIEG ARTHUR M (US)
BARSOUM JAMES (US)
Application Number:
PCT/US2014/059111
Publication Date:
April 09, 2015
Filing Date:
October 03, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
RANA THERAPEUTICS INC (US)
International Classes:
C12N15/63; A61K31/7088; A61P21/00; C12N5/0793
Foreign References:
US20100087511A12010-04-08
US20090324549A12009-12-31
US20110086833A12011-04-14
Other References:
CORCIA, P. ET AL.: "The importance of the SMN genes in the genetics of sporadic ALS", AMYOTROPHIC LATERAL SCLEROSIS, vol. 10, no. 5-6, December 2009 (2009-12-01), pages 436 - 440, XP008183189
TSUIJI, H. ET AL.: "Spliceosome integrity is defective in the motor neuron diseases ALS and SMA", EMBO MOL. MED., vol. 5, no. 2, 25 January 2013 (2013-01-25), pages 221 - 234, XP055331692
See also references of EP 3052632A4
Attorney, Agent or Firm:
YOUNG, Daniel, W. (Greenfield & Sacks P.C.,600 Atlantic Avenu, Boston MA, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method of increasing expression of SMN in a cell of a subject having ALS, the method comprising delivering to the cell a first single stranded oligonucleotide comprising a region of complementarity that is complementary with at least 8 consecutive nucleotides of a PRC2- associated region of an SMN gene.

2. The method of claim 1, wherein the cell comprises a genetic alteration associated with the ALS in SOD1, ALS2, SETX, FUS/TLS, VAPB, ANG, TDP-43, FIG4, OPTN, ATXN2, VCP, UBQLN2, SIGMAR1, CHMP2B, PFNl.or C9orf72

3. The method of claim 1 or 2, wherein the cell comprises a genetic alteration associated with the ALS in SOD1.

4. The method of any one of claims 1-3, wherein the cell is a motor neuron.

5. The method of any one of claims 1-4, wherein the first single stranded oligonucleotides comprises a sequence that is 5'X-Y-Z, wherein X is any nucleotide and wherein X is anchored at the 5' end of the oligonucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a human seed sequence of a microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length.

6. The method of any one of claims 1-5, wherein the first single stranded oligonucleotide is 8 to 30 nucleotides in length.

7. The method of any one of claims 1-6, wherein the method further comprises administering a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN2.

8. The method of claim 7, wherein the splice control sequence resides in an exon of SMN2.

9. The method of claim 8, wherein the exon is exon 7 or exon 8.

10. The method of claim 7, wherein the splice control sequence traverses an intron-exon junction of SMN2.

11. The method of claim 10, wherein the intron-exon junction is the intron 6/exon 7 junction or the intron 7/exon 8 junction.

12. The method of claim 7, wherein the splice control sequence resides in an intron of SMN2.

13. The method of claim 12, wherein the intron is intron 6 or intron 7.

14. The method of any one of claims 7-13, wherein the first single stranded oligonucleotide and the second single stranded oligonucleotide are administered

simultaneously.

15. The method of any one of claims 7-14, wherein the first single stranded oligonucleotide is covalently linked to the second single stranded oligonucleotide through a linker. 16. The method of claim 15, wherein the linker comprises an oligonucleotide, a peptide, a low pH-labile bond, or a disulfide bond.

17. The method of any one of claims 7-16, wherein at least one nucleotide of the first or second single stranded oligonucleotide is a nucleotide analogue.

18. The method of claim 7-17, wherein the first single stranded oligonucleotide or second single stranded oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide.

19 The method of any one of claims 7-18, wherein the second single stranded oligonucleotide is 8 to 30 nucleotides in length.

20. The method of any one of claims 1-19, wherein the cell comprises an SMN1 gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA).

21. A method increasing levels of SMN in a subject having ALS, the method comprising administering a first single stranded oligonucleotide comprising a region of complementarity that is complementary with at least 8 consecutive nucleotides of a PRC2- associated region of an SMN gene to the subject. 22. A method of treating ALS, the method comprising administering a first single stranded oligonucleotide comprising a region of complementarity that is complementary with at least 8 consecutive nucleotides of a PRC2-associated region of an SMN gene to the subject.

23. The method of claim 21 or 22, wherein the subject has a mutation in a gene selected from SOD1, FUS/TLS, or TDP-43.

24. The method of any one of claims 21-23, wherein the single stranded oligonucleotide is administered intrathecally. 25. The method of any one of claims 21-24, wherein the first single stranded oligonucleotides comprises a sequence that is 5'X-Y-Z, wherein X is any nucleotide and wherein X is anchored at the 5' end of the oligonucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a human seed sequence of a microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length. 26. The method of any one of claims 21-25, wherein the first single stranded oligonucleotide is 8 to 30 nucleotides in length.

27. The method of any one of claims 21-26, wherein at least one nucleotide of the first single stranded oligonucleotide is a nucleotide analogue.

28. The method of any one of claims 21-27, wherein the method further comprises administering a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN2.

29. The method of claim 28, wherein the splice control sequence resides in an exon of SMN2.

30. The method of claim 29, wherein the exon is exon 7 or exon 8. 31. The method of claim 28, wherein the splice control sequence traverses an intron-exon junction of SMN2.

32. The method of claim 31, wherein the intron-exon junction is the intron 6/exon 7 junction or the intron 7/exon 8 junction.

33. The method of claim 28, wherein the splice control sequence resides in an intron of SMN2.

34. The method of claim 33, wherein the intron is intron 6 or intron 7.

35. The method of any one of claims 28-34, wherein the first single stranded oligonucleotide and the second single stranded oligonucleotide are administered

simultaneously.

36. The method of any one of claims 28-35, wherein the first single stranded oligonucleotide is covalently linked to the second single stranded oligonucleotide through a linker.

37. The method of claim 36, wherein the linker comprises an oligonucleotide, a peptide, a low pH-labile bond, or a disulfide bond.

38. The method of any one of claims 28-37, wherein at least one nucleotide of the second single stranded oligonucleotide is a nucleotide analogue.

39. The method of claim 28-38, wherein the first single stranded oligonucleotide or second single stranded oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide.

40. The method of any one of claims 38-39, wherein the second single stranded oligonucleotide is 8 to 30 nucleotides in length.

41. A method for promoting Gem formation in cells having a spliceosome defect, the method comprising delivering to the cells a single stranded oligonucleotide comprising a region of complementarity that is complementary with at least 8 consecutive nucleotides of a PRC2- associated region of an SMN gene.

42. The method of claim 41, wherein the cell is a motor neuron of a patient having a motor neuron disease.

43. The method of claim 42, wherein the motor neuron disease is ALS or SMA.

44. The method of any one of claims 41 to 43 further comprising evaluating spliceosome integrity in the cells prior to and/or following delivery of the single stranded oligonucleotide to the cells.

45. The method of claim 44, wherein evaluating spliceosome intergrity comprises determining the localization of FUS, TDP-43, SMN, Gemin3, Gemin4 or another Gem marker in the nuclei of the cells.

46. The method of any one of claims 41-45, wherein the cells comprise an SMN1 gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA).

Description:
COMPOSITIONS AND METHODS FOR TREATING AMYOTROPHIC LATERAL

SCLEROSIS

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 61/887,019, entitled "COMPOSITIONS AND METHODS FOR

TREATING AMYOTROPHIC LATERAL SCLEROSIS", filed October 4, 2013, the contents of which are incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

The invention relates to oligonucleotide based compositions, as well as methods of using oligonucleotide based compositions for treating disease. BACKGROUND OF THE INVENTION

Amyotrophic Lateral Sclerosis (ALS) is a progressive neurodegenerative disease that affects motor neurons and eventually results in death. A number of genes have been associated with ALS, including superoxide dismutase 1 (SOD1), fused in

sarcoma/translocated in liposarcoma (FUS/TLS), and to the loss of normal TAR DNA- binding protein 43 (TDP-43), survival of motor neuron (SMN) and others. There is currently no cure for ALS.

Survival of motor neuron (SMN) is a protein involved in transcriptional splicing through its involvement in assembly of small nuclear ribonucleoproteins (snRNPs). snRNPs are protein-RNA complexes that bind with pre-mRNA to form a spliceosome, which then splices the pre-mRNA, most often resulting in removal of introns. Three genes encode SMN or a variant of SMN, including SMNl (survival of motor neuron 1, telomeric), SMN2 (survival of motor neuron 2, centromeric), and SMNP (survival of motor neuron 1, telomeric pseudogene). SMNl and SMN2 are a result of a gene duplication at 5ql3 in humans. A lack of SMN activity results in widespread splicing defects, especially in spinal motor neurons, and degeneration of the spinal cord lower motor neurons.

SUMMARY OF THE INVENTION Aspects of the invention relate to methods and compositions for treating motor neuron diseases, such as, Amyotrophic Lateral Sclerosis (ALS), Primary Lateral Sclerosis,

Progressive Muscular Atrophy, Progressive Bulbar Palsy or Pseudobulbar Palsy. In some embodiments, methods are provided for treating ALS that involve administering to a subject having ALS one or more single stranded oligonucleotides that cause upregulation of SMN in cells (e.g., motor neurons). In some embodiments, single stranded oligonucleotides are provided that target a PRC2-associated region of a SMN gene (e.g., human SMNl, human SMN2) and thereby cause upregulation of the gene. For example, according to some aspects of the invention methods are provided for increasing expression of full-length SMN protein in a cell for purposes of treating ALS. In other aspects, methods and compositions are provided herein for promoting Gem formation in cells (e.g., motor neurons) of patients having a motor neuron disease (e.g., ALS, Primary Lateral Sclerosis, Progressive Muscular Atrophy, Progressive Bulbar Palsy, Pseudobulbar Palsy, or SMA). Accordingly, in some embodiments, methods and compositions are provided for improving, at least partially, spliceosome integrity in neurons.

Accordingly, aspects of the invention disclosed herein provide methods and compositions that are useful for upregulating SMN (SMNl, SMN2) in cells. In some embodiments, single stranded oligonucleotides are provided that target a PRC2-associated region of the gene encoding SMNl or SMN2. In some embodiments, these single stranded oligonucleotides activate or enhance expression of SMNl or SMN2 by relieving or preventing PRC2 mediated repression of SMNl or SMN2. In some embodiments, the cells comprise an SMNl gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA).

In some embodiments, methods provided herein comprise delivering to a cell a first single stranded oligonucleotide complementary with a PRC2-associated region of an SMN gene, e.g., a PRC2-associated region of SMNl or SMN2, and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of an SMN gene, e.g., a precursor mRNA of SMNl or SMN2, in amounts sufficient to increase expression of a mature mRNA of SMNl or SMN2 that comprises (or includes) exon 7 in the cell. In some embodiments, the cell comprises an SMNl gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA).

According to some aspects of the invention single stranded oligonucleotides are provided that have a region of complementarity that is complementarty with (e.g., at least 8 consecutive nucleotides of ) a PRC2-associated region of an SMN gene, e.g., a PRC2- associated region of the nucleotide sequence set forth as SEQ ID NO: 1, 2, 4, or 5. In some embodiments, the oligonucleotide has at least one of the following features: a) a sequence that is 5 'X-Y-Z, in which X is any nucleotide and in which X is at the 5' end of the oligonucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a human seed sequence of a microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length; b) a sequence that does not comprise three or more consecutive guanosine

nucleotides; c) a sequence that has less than a threshold level of sequence identity with every sequence of nucleotides, of equivalent length to the second nucleotide sequence, that are between 50 kilobases upstream of a 5 '-end of an off-target gene and 50 kilobases downstream of a 3 '-end of the off-target gene; d) a sequence that is complementary to a PRC2-associated region that encodes an RNA that forms a secondary structure comprising at least two single stranded loops; and e) a sequence that has greater than 60% G-C content. In some

embodiments, the single stranded oligonucleotide has at least two of features a), b), c), d), and e), each independently selected. In some embodiments, the single stranded

oligonucleotide has at least three of features a), b), c), d), and e), each independently selected. In some embodiments, the single stranded oligonucleotide has at least four of features a), b), c), d), and e), each independently selected. In some embodiments, the single stranded oligonucleotide has each of features a), b), c), d), and e). In certain embodiments, the oligonucleotide has the sequence 5 'X-Y-Z, in which the oligonucleotide is 8-50 nucleotides in length.

According to some aspects of the invention, single stranded oligonucleotides are provided that have a sequence X-Y-Z, in which X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length, in which the single stranded oligonucleotide is complementary with a PRC2- associated region of an SMN gene, e.g., a PRC2-associated region of the nucleotide sequence set forth as SEQ ID NO: 1, 2, 4, or 5. In some aspects of the invention, single stranded oligonucleotides are provided that have a sequence 5' -X-Y-Z, in which X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length, in which the single stranded oligonucleotide is complementary with at least 8 consecutive nucleotides of a PRC2-associated region of an SMN gene, e.g., a PRC2- associated region of the nucleotide sequence set forth as SEQ ID NO: 1, 2, 4, or 5. In some embodiments, Y is a sequence selected from Table 1. In some embodiments, the PRC2-associated region is a sequence listed in any one of SEQ ID NOS: 9 to 18.

In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 13087 or 13108 to 13116, or a fragment thereof that is at least 8 nucleotides. In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 13087 or 13108 to 13116, in which the 5' end of the nucleotide sequence provided is the 5' end of the oligonucleotide. In some embodiments, the region of complementarity (e.g., the at least 8 consecutive nucleotides) is also present within the nucleotide sequence set forth as SEQ ID NO: 7 or 8.

In some embodiments, a PRC2-associated region is a sequence listed in any one of SEQ ID NOS: 9 to 14. In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094, wherein the 5' end of the nucleotide sequence provided is the 5' end of the oligonucleotide. In some embodiments, the at least 8

consecutive nucleotides are also present within the nucleotide sequence set forth as SEQ ID NO: 7.

In some embodiments, a PRC2-associated region is a sequence listed in any one of SEQ ID NOS: 15 to 18. In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916- 3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087 and 13108- 13116 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the at least 8 consecutive nucleotides are present within the nucleotide sequence set forth as SEQ ID NO: 8. In some embodiments, the single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 13087 or 13108 to 13116. In some embodiments, the oligonucleotide is up to 50 nucleotides in length. In some embodiments, the single stranded oligonucleotide comprises a fragment of at least 8 nucleotides of a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 13087 or 13108 to 13116.

In some embodiments, a single stranded oligonucleotide comprises a nucleotide sequence as set forth in Table 4. In some embodiments, the single stranded oligonucleotide comprises a fragment of at least 8 nucleotides of a nucleotide sequence as set forth in Table 4 or Table 6. In some embodiments, a single stranded oligonucleotide consists of a nucleotide sequence as set forth in Table 4 or Table 6.

According to some aspects of the invention, compounds are provided that comprise the general formula A-B-C, wherein A is a single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of a gene, B is a linker, and C is a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, B comprises an oligonucleotide, peptide, low pH labile bond, or disulfide bond. In some embodiments, the splice control sequence resides in an exon of the gene. In some embodiments, the splice control sequence traverses an intron-exon junction of the gene. In some embodiments, the splice control sequence resides in an intron of the gene. In some embodiments, the splice control sequence comprises at least one hnRNAP binding sequence. In some embodiments, hybridization of an oligonucleotide having the sequence of C with the splice control sequence of the precursor mRNA in a cell results in inclusion of a particular exon in a mature mRNA that arises from processing of the precursor mRNA in the cell. In some embodiments, hybridization of an oligonucleotide having the sequence of C with the splice control sequence of the precursor mRNA in a cell results in exclusion of a particular intron or exon in a mature mRNA that arises from processing of the precursor mRNA in the cell.

In some embodiments, the gene is SMNl or SMN2. In some embodiments, the splice control sequence resides in intron 6, intron 7, exon 7, exon 8 or at the junction of intron 7 and exon 8 of SMNl or SMN2. In some embodiments, the splice control sequence comprises the sequence: CAGCAUUAUGAAAG (SEQ ID NO: 13100). In some embodiments, B comprises a sequence selected from: TCACTTTCATAATGCTGG (SEQ ID NO: 13088); TCACTTTCATAATGC (SEQ ID NO: 13089); CACTTTCATAATGCT (SEQ ID NO: 13090); ACTTTCATAATGCTG (SEQ ID NO: 13090); and CTTTCATAATGCTGG (SEQ ID NO: 13092).

In some embodiments, A has a sequence 5'-X-Y-Z, wherein X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1-23 nucleotides in length. In some embodiments, the PRC2-associated region of an SMN2 gene is a PRC2-associated region within SEQ ID NO: 1, 2, 4 or 5. In some embodiments, Y is a sequence selected from Table 1. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 9 to 23. In some embodiments, A comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13088 to 13094 or a fragment thereof that is at least 8 nucleotides. In some embodiments, A comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13088 to 13094, wherein the 5' end of the nucleotide sequence provided is the 5' end of A. In some embodiments, the at least 8 consecutive nucleotides are also present within the nucleotide sequence set forth as SEQ ID NO: 7. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 24 to 29.

In some embodiments, A comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, and 13108-13116 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the at least 8 consecutive nucleotides are present within the nucleotide sequence set forth as SEQ ID NO: 8. In some embodiments, A does not comprise three or more consecutive guanosine nucleotides. In some embodiments, A does not comprise four or more consecutive guanosine nucleotides. In some embodiments, A or C is 8 to 30 nucleotides in length. In some embodiments, A is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the PRC2-associated region are cytosine or guanosine nucleotides. In some embodiments, B is an oligonucleotide comprising 1 to 10 thymidines or uridines. In some embodiments, B is more susceptible to cleavage in a mammalian extract than A and C.

In some embodiments, A comprises a nucleotide sequence selected from

GCTUTGGGAAGUAUG (SEQ ID NO: 11394), CUTUGGGAAGTATG (SEQ ID NO: 11395) and GGTACATGAGTGGCT (SEQ ID NO: 11419); B comprises the nucleotide sequence TTTT or UUUU; and C comprises the nucleotide sequence

TCACTTTCATAATGCTGG (SEQ ID NO: 13088); TCACTTTCATAATGC (SEQ ID NO: 13089); CACTTTCATAATGCT (SEQ ID NO: 13090); ACTTTCATAATGCTG (SEQ ID NO: 13091); or CTTTCATAATGCTGG (SEQ ID NO: 13092), and wherein the 3' end of A is linked to the 5' end of B, and the 3' end of B is linked to 5' end of C.

In some embodiments, the single stranded oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the single stranded oligonucleotide does not comprise four or more consecutive guanosine nucleotides.

In some embodiments, the single stranded oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the single stranded oligonucleotide is up to 50 nucleotides in length. In some embodiments, the single stranded oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the PRC2- associated region are cytosine or guanosine nucleotides.

In some embodiments, the single stranded oligonucleotide is complementary with at least 8 consecutive nucleotides of a PRC2-associated region of an SMN gene, e.g., a PRC2- associated region of a nucleotide sequence set forth as SEQ ID NO: 1, 2, 4, or 5, in which the nucleotide sequence of the single stranded oligonucleotide comprises one or more of a nucleotide sequence selected from the group consisting of

(a) (X)Xxxxxx, (X)xXxxxx, (X)xxXxxx, (X)xxxXxx, (X)xxxxXx and (X)xxxxxX, (b) (X)XXxxxx, (X)XxXxxx, (X)XxxXxx, (X)XxxxXx, (X)XxxxxX, (X)xXXxxx,

(X)xXxXxx, (X)xXxxXx, (X)xXxxxX, (X)xxXXxx, (X)xxXxXx, (X)xxXxxX, (X)xxxXXx, (X)xxxXxX and (X)xxxxXX,

(c) (X)XXXxxx, (X)xXXXxx, (X)xxXXXx, (X)xxxXXX, (X)XXxXxx, (X)XXxxXx, (X)XXxxxX, (X)xXXxXx, (X)xXXxxX, (X)xxXXxX, (X)XxXXxx, (X)XxxXXx

(X)XxxxXX, (X)xXxXXx, (X)xXxxXX, (X)xxXxXX, (X)xXxXxX and (X)XxXxXx,

(d) (X)xxXXX, (X)xXxXXX, (X)xXXxXX, (X)xXXXxX, (X)xXXXXx,

(X)XxxXXXX, (X)XxXxXX, (X)XxXXxX, (X)XxXXx, (X)XXxxXX, (X)XXxXxX, (X)XXxXXx, (X)XXXxxX, (X)XXXxXx, and (X)XXXXxx,

(e) (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXXxX and (X)XXXXXx, and

(f) XXXXXX, XxXXXXX, XXxXXXX, XXXxXXX, XXXXxXX, XXXXXxX and XXXXXXx, wherein "X" denotes a nucleotide analogue, (X) denotes an optional nucleotide analogue, and "x" denotes a DNA or RNA nucleotide unit.

In some embodiments, at least one nucleotide of the oligonucleotide is a nucleotide analogue. In some embodiments, the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5 °C compared with an oligonucleotide that does not have the at least one nucleotide analogue.

In some embodiments, at least one nucleotide of the oligonucleotide comprises a 2' O-methyl. In some embodiments, each nucleotide of the oligonucleotide comprises a 2' O- methyl. In some embodiments, the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide. In some embodiments, the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide. In some embodiments, each nucleotide of the oligonucleotide is a LNA nucleotide.

In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2'-fluoro-deoxyribonucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2'-0- methyl nucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides. In some embodiments, the 5' nucleotide of the oligonucleotide is a

deoxyribonucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2' -O-methyl nucleotides. In some embodiments, the 5' nucleotide of the oligonucleotide is a LNA nucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5' and 3' ends of the deoxyribonucleotides.

In some embodiments, the single stranded oligonucleotide comprises modified internucleotide linkages (e.g., phosphorothioate internucleotide linkages or other linkages) between at least two, at least three, at least four, at least five or more nucleotides. In some embodiments, the single stranded oligonucleotide comprises modified internucleotide linkages (e.g., phosphorothioate internucleotide linkages or other linkages) between between all nucleotides.

In some embodiments, the nucleotide at the 3' position of the oligonucleotide has a 3' hydroxyl group. In some embodiments, the nucleotide at the 3' position of the

oligonucleotide has a 3' thiophosphate. In some embodiments, the single stranded oligonucleotide has a biotin moiety or other moiety conjugated to its 5' or 3' nucleotide. In some embodiments, the single stranded oligonucleotide has cholesterol, Vitamin A, folate, sigma receptor ligands, aptamers, peptides, such as CPP, hydrophobic molecules, such as lipids, ASGPR or dynamic polyconjugates and variants thereof at its 5' or 3' end.

According to some aspects of the invention compositions are provided that comprise any of the oligonucleotides disclosed herein, and a carrier. In some embodiments, compositions are provided that comprise any of the oligonucleotides in a buffered solution. In some embodiments, the oligonucleotide is conjugated to the carrier. In some embodiments, the carrier is a peptide. In some embodiments, the carrier is a steroid. According to some aspects of the invention pharmaceutical compositions are provided that comprise any of the oligonucleotides disclosed herein, and a pharmaceutically acceptable carrier.

According to other aspects of the invention, kits are provided that comprise a container housing any of the compositions disclosed herein.

According to some aspects of the invention, methods of increasing expression of SMNl or SMN2 in a cell are provided. In some embodiments, the cell comprises a SMNl gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA). In some embodiments, the cell has a wild- type SMNl gene. In some embodiments, the methods involve delivering any one or more of the single stranded oligonucleotides disclosed herein into the cell. In some embodiments, delivery of the single stranded oligonucleotide into the cell results in a level of expression of SMNl or SMN2 that is greater (e.g., at least 50% greater) than a level of expression of SMNl or SMN2 in a control cell that does not comprise the single stranded oligonucleotide.

According to some aspects of the invention, methods of increasing levels of SMNl or SMN2 in a subject are provided. According to some aspects of the invention, methods of treating a condition (e.g., ALS, Primary Lateral Sclerosis, Progressive Muscular Atrophy, Progressive Bulbar Palsy, or Pseudobulbar Palsy) associated with decreased levels of SMNl or SMN2 in a subject are provided. In some embodiments, the methods involve

administering any one or more of the single stranded oligonucleotides disclosed herein to the subject.

Aspects of the invention relate to methods of increasing expression of SMN protein in a cell. In some embodiments, the cell comprises an SMNl gene that does not have a mutation associated with Spinal Muscular Atrophy (SMA). In some embodiments, the method comprise delivering to the cell a first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN2, in amounts sufficient to increase expression of a mature mRNA of SMN2 that comprises exon 7 in the cell. In some embodiments, the region of complementarity with at least 8 consecutive nucleotides of a PRC2-associated region of SMN 2 has at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, or more mismatches with a corresponding region of SMN1. As used herein the term, "splice control sequence" refers to a nucleotide sequence that when present in a precursor mRNA influences splicing of that precursor mRNA in a cell. In some embodiments, a splice control sequence includes one or more binding sites for a molecule that regulates mRNA splicing, such as a hnRNAP protein. In some embodiments, a splice control sequence comprises the sequence CAG or AAAG. In some embodiments, a splice control sequence resides in an exon (e.g., an exon of SMN1 or SMN2, such as exon 7 or exon 8). In some embodiments, a splice control sequence traverses an intron-exon junction (e.g., an intron-exon junction of SMN1 or SMN2, such as the intron 6/exon 7 junction or the intron 7/exon 8 junction). In some embodiments, a splice control sequence resides in an intron (e.g., an intron of SMN1 or SMN2, such as intron 6 or intron 7). In some embodiments, a splice control sequence comprises the sequence:

CAGCAUUAUGAAAG (SEQ ID NO: 13100) or a portion thereof.

In some embodiments, the second single stranded oligonucleotide is splice switching oligonucleotide that comprises a sequence selected from: TCACTTTCATAATGCTGG (SEQ ID NO: 13088); TCACTTTCATAATGC (SEQ ID NO: 13089);

CACTTTCATAATGCT (SEQ ID NO: 13090); ACTTTCATAATGCTG (SEQ ID NO:

13091); and CTTTCATAATGCTGG (SEQ ID NO: 13092). In some embodiments, the second single stranded oligonucleotide is 8 to 30 nucleotides in length.

In some embodiments, the first single stranded oligonucleotide has a sequence 5'-X- Y-Z, wherein X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1-23 nucleotides in length. In some embodiments, the PRC2-associated region of an SMN2 gene is a PRC2-associated region within SEQ ID NO: 1, 2, 4 or 5. In some embodiments, Y is a sequence selected from Table 1. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 9 to 23. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13088 to 13094 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13088 to 13094, wherein the 5' end of the nucleotide sequence provided is the 5' end of the first single stranded oligonucleotide. In some embodiments, the at least 8 consecutive nucleotides are also present within the nucleotide sequence set forth as SEQ ID NO: 7.

In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 24 to 29. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439- 3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, and 13108-13116 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the at least 8 consecutive nucleotides are present within the nucleotide sequence set forth as SEQ ID NO: 8. In some embodiments, the first single stranded oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the first single stranded oligonucleotide does not comprise four or more consecutive guanosine nucleotides. In some embodiments, the first single stranded oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the first single stranded oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the PRC2-associated region are cytosine or guanosine nucleotides.

In some embodiments, the first single stranded oligonucleotide and the second single stranded oligonucleotide are delivered to the cell simultaneously. In some embodiments, the cell is in a subject and the step of delivering to the cell comprises administering the first single stranded oligonucleotide and the second single stranded oligonucleotide to the subject as a co-formulation. In some embodiments, the first single stranded oligonucleotide is covalently linked to the second single stranded oligonucleotide through a linker. In some embodiments, the linker comprises an oligonucleotide, a peptide, a low pH-labile bond, or a disulfide bond. In some embodiments, the linker comprises an oligonucleotide, optionally wherein the oligonucleotide comprises 1 to 10 thymidines or uridines. In some embodiments, the linker is more susceptible to cleavage in a mammalian extract than the first and second single stranded oligonucleotides. In some embodiments, the first single stranded oligonucleotide is not covalently linked to the second single stranded oligonucleotide. In some embodiments, the first single stranded oligonucleotide and the second single stranded oligonucleotide are delivered to the cell separately.

According to some aspects of the invention, methods are provided for treating ALS in a subject. The methods, in some embodiments, comprise administering to the subject a first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of SMN2 and a second single stranded oligonucleotide

complementary with a splice control sequence of a precursor mRNA of SMN2, in amounts sufficient to increase expression of SMN protein in the subject.

According to some aspects of the invention methods are provided for treating ALS in a subject that involve administering to the subject a first single stranded oligonucleotide complementary with a PRC2-associated region of SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN2, in amounts sufficient to increase expression of SMN protein in the subject. Related compositions are also provided. In some embodiments, compositions are provided that comprise a first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2- associated region of SMN2, and a second single stranded

oligonucleotide complementary to a splice control sequence of a precursor mRNA of SMN2. In some embodiments, compositions are provided that comprise a single stranded

oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of a gene, linked via a linker to a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. Related kits comprising single stranded oligonucleotides that regulate SMN1 or SMN2 expression are also provided.

According to some aspects of the invention compositions are provided that comprise any of the oligonucleotides or compounds disclosed herein, and a carrier. In some embodiments, compositions are provided that comprise any of the oligonucleotides or compounds in a buffered solution. In some embodiments, the oligonucleotide is conjugated to the carrier. In some embodiments, the carrier is a peptide. In some embodiments, the carrier is a steroid. According to some aspects of the invention pharmaceutical compositions are provided that comprise any of the oligonucleotides disclosed herein, and a pharmaceutically acceptable carrier. According to some aspects of the invention, compositions are provided that comprise a first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of SMN2, and a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of SMN2. In some embodiments, the splice control sequence resides in an exon of SMN2. In some

embodiments, the exon is exon 7 or exon 8. In some embodiments, the splice control sequence traverses an intron-exon junction of SMN2. In some embodiments, the intron-exon junction is the intron 6/exon 7 junction or the intron 7/exon 8 junction. In some

embodiments, the splice control sequence resides in an intron of SMN2. In some

embodiments, the intron is intron 6 or intron 7 (SEQ ID NO: 13101). In some embodiments, the splice control sequence comprises the sequence: CAGCAUUAUGAAAG (SEQ ID NO: 13100) or a portion thereof. In some embodiments, the splice control sequence comprises at least one hnRNAP binding sequence. In some embodiments, the second single stranded oligonucleotide comprises a sequence selected from: TCACTTTCATAATGCTGG (SEQ ID NO: 13088); TCACTTTCATAATGC (SEQ ID NO: 13089); CACTTTCATAATGCT (SEQ ID NO: 13090); ACTTTCATAATGCTG (SEQ ID NO: 13091); and

CTTTCATAATGCTGG (SEQ ID NO: 13092). In some embodiments, the first single stranded oligonucleotide has a sequence 5'-X-Y-Z, wherein X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1-23 nucleotides in length. In some

embodiments, the PRC2-associated region of SMN2 is a PRC2-associated region within SEQ ID NO: 1,2, 4 or 5. In some embodiments, Y is a sequence selected from Table 1. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 9 to 23. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094, wherein the 5' end of the nucleotide sequence provided is the 5' end of the first single stranded oligonucleotide. In some embodiments, the at least 8 consecutive nucleotides are also present within the nucleotide sequence set forth as SEQ ID NO: 7. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 24 to 29. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, and 13108-13116 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the at least 8 consecutive nucleotides are present within the nucleotide sequence set forth as SEQ ID NO: 8. In some embodiments, the first single stranded oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the first single stranded oligonucleotide does not comprise four or more consecutive guanosine nucleotides. In some embodiments, the first and/or second single stranded oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the first single stranded oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the PRC2-associated region are cytosine or guanosine nucleotides. In some embodiments, the first single stranded oligonucleotide is covalently linked to the second single stranded oligonucleotide through a linker. In some embodiments, the linker comprises an oligonucleotide, a peptide, a low pH-labile bond, or a disulfide bond. In some embodiments, the linker comprises an oligonucleotide, optionally wherein the oligonucleotide comprises 1 to 10 thymidines or uridines. In some embodiments, the linker is more susceptible to cleavage in a mammalian extract than the first and second single stranded oligonucleotides. In some embodiments, the first single stranded

oligonucleotide is not covalently linked to the second single stranded oligonucleotide. In some embodiments, the composition further comprises a carrier. In some embodiments, the carrier is a pharmaceutically acceptable carrier.

Further aspects of the invention provide methods for selecting oligonucleotides for activating or enhancing expression of SMNl or SMN2. In some embodiments, methods are provided for selecting a set of oligonucleotides that is enriched in candidates (e.g., compared with a random selection of oligonucleotides) for activating or enhancing expression of SMNl or SMN2. Accordingly, the methods may be used to establish sets of clinical candidates that are enriched in oligonucleotides that activate or enhance expression of SMNl or SMN2. Such libraries may be utilized, for example, to identify lead oligonucleotides for developing therapeutics to treat SMNl or SMN2. Furthermore, in some embodiments, oligonucleotide chemistries are provided that are useful for controlling the pharmacokinetics, biodistribution, bioavailability and/or efficacy of the single stranded oligonucleotides for activating expression of SMN1 or SMN2.

According to other aspects of the invention, kits are provided that comprise a container housing any of the compositions disclosed herein. According to other aspects of the invention, kits are provided that comprise a first container housing first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2-associated region of a gene; and a second container housing a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, the splice control sequence resides in an exon of the gene. In some

embodiments, the splice control sequence traverses an intron-exon junction of the gene. In some embodiments, the splice control sequence resides in an intron of the gene. In some embodiments, the splice control sequence comprises at least one hnRNAP binding sequence. In some embodiments, hybridization of an oligonucleotide having the sequence of C with the splice control sequence of the precursor mRNA in a cell results in inclusion of a particular exon in a mature mRNA that arises from processing of the precursor mRNA in the cell. In some embodiments, hybridization of an oligonucleotide having the sequence of C with the splice control sequence of the precursor mRNA in a cell results in exclusion of a particular intron or exon in a mature mRNA that arises from processing of the precursor mRNA in the cell. In some embodiments, the gene is SMN1 or SMN2. In some embodiments, the splice control sequence resides in intron 6, intron 7, exon 7, exon 8 or at the junction of intron 7 and exon 8. In some embodiments, the splice control sequence comprises the sequence:

CAGCAUUAUGAAAG (SEQ ID NO: 13100). In some embodiments, the second single stranded oligonucleotide comprises a sequence selected from: TCACTTTCATAATGCTGG (SEQ ID NO: 13088); TCACTTTCATAATGC (SEQ ID NO: 13089);

CACTTTCATAATGCT (SEQ ID NO: 13090); ACTTTCATAATGCTG (SEQ ID NO: XX); and CTTTCATAATGCTGG (SEQ ID NO: 13091). In some embodiments, the first single stranded oligonucleotide has a sequence 5'-X-Y-Z, wherein X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a seed sequence of a human microRNA, and Z is a nucleotide sequence of 1-23 nucleotides in length. In some embodiments, the PRC2-associated region of an SMN2 gene is a PRC2-associated region within SEQ ID NO: 1, 2, 4 or 5. In some embodiments, Y is a sequence selected from Table 1. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 9 to 23. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 30 to 8329 and 13093 to 13094, wherein the 5' end of the nucleotide sequence provided is the 5' end of the first single stranded oligonucleotide. In some embodiments, the at least 8 consecutive nucleotides are also present within the nucleotide sequence set forth as SEQ ID NO: 7. In some embodiments, the PRC2-associated region is a sequence set forth in any one of SEQ ID NOS: 24 to 29. In some embodiments, the first single stranded oligonucleotide comprises a nucleotide sequence as set forth in any one of SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, and 13108-13116 or a fragment thereof that is at least 8 nucleotides. In some embodiments, the at least 8 consecutive nucleotides are present within the nucleotide sequence set forth as SEQ ID NO: 8.

BRIEF DESCRIPTION OF DRAWINGS FIG. 1 provides a schematic of SMN1 and SMN2 mRNA processing

FIG. 2 provides a table outlining genotypes and patent information of cell lines tested in Example 2. Baseline SMN protein levels in the cell lines are also depicted.

FIG. 3 depicts results of RT-PCR assays showing effects on SMN mRNA expression of oligonucleotides directed against a PRC2-associated region of SMN2 (oligos 1-52 and 59- 101) and splice switching oligonucleotides (oligos 53-58) (PCR primers directed against exon 1 of SMN1/2.) in cell line 3814.

FIG. 4 depicts results of RT-PCR assays showing effects on SMN mRNA expression of oligonucleotides directed against a PRC2-associated region of SMN2 (oligos 1-52 and 59- 101) and splice switching oligonucleotides (oligos 53-58) (PCR primers directed against exon 1 of SMN1/2.) in cell line 3813.

FIG. 5 shows that splice switching oligonucleotides (oligoes 53-58) increase expression of full length SMN2. Results are based on a gel separation analysis of PCR products obtained following a Ddel restriction digest. Two cell lines were tested, 3813 and 9677. Oligo 84, which targets a PRC2-associated region of SMN2, did not exhibit an increase in full length SMN2 expression when delivered alone to cells. FIG. 6 provides results of an SMN ELISA (Enzo) showing that certain oligonucleotides directed against a PRC2-associated region of SMN2 alone do not significantly increase SMN2 protein 24h post-transfection in certain patient fibroblasts with reduced SMN expression (compared to Lipofectamine treated cells - dashed line).

FIG. 7 provides results of an SMN ELISA showing that oligonucleotides directed against a PRC2-associated region of SMN2 in combination with a splice switching oligonucleotide (oligo 53) significantly increase SMN2 protein 24h post-transfection in patient fibroblasts with reduced SMN expression (compared to Lipofectamine treated cells - dashed line).

FIG. 8 provides results of an SMN ELISA showing that oligonucleotides directed against a PRC2-associated region of SMN2 in combination with a splice switching oligonucleotide (oligo 54) significantly increase SMN2 protein 24h post-transfection in patient fibroblasts with reduced SMN expression (compared to Lipofectamine treated cells - dashed line).

FIG. 9 provides results of an RT-PCR assay showing that oligonucleotides directed against a PRC2-associated region of SMN2 in combination with a splice switching oligonucleotide (oligo 53 or 54) significantly increase SMN2 protein 24h post-transfection in SMA patient fibroblasts (compared to negative control oligo and Lipofectamine treated cells). LNA/2'OMe alternating oligonucleotide (LM design) and DNA/LNA alternating

oligonucleotides (DL design) were tested.

FIG. 10A provides a graph showing levels of EED (Embryonic ectoderm

development) mRNA after EED knockdown.

FIG. 10B provides a graph showing levels of SMN2 mRNA after EED knockdown. Levels of full-length (FL) and the exon 7 deleted (del7) SMN2 mRNA levels are shown.

FIG. IOC provides a graph showing levels of SMN2 mRNA after treatment with SMN oligos 77 or 83 compared to controls (cells only or cell treated with lipofectamine only). Levels of full-length (FL) and the exon 7 deleted (del7) SMN2 mRNA levels are shown.

FIG. 11 A provides a diagram showing the SMN2 locus and the locations of certain regions within the locus that are shown in FIGs. 11-14.

FIGs. 1 IB and 11C provide a series of graphs showing that knockdown of PRC2 components reduces the presence of EZH2 in the SMN chromatin. FIG. 1 IB shows EZH2 ChIP (chromatin immunoprecipitation) data and FIG. 11C shows H3K27m3 ChIP data. FIG. 1 ID provides a graph showing the presence of H3, H3K4m3, H3K27m3, H3K36m3, RNA polII_S2 and EZH2 at the HOXC13 promoter.

FIGs. 12A-D provide a series of graphs showing that knockdown of PRC2

components leads to an increase in marks of transcriptional activity. FIG. 12A shows H3K36m3 ChIP data, FIG. 12B shows RNA Pol II S2 ChIP data, FIG. 12. C shows H3K4m3 ChIP data, and FIG. 12D shows H3 ChIP data.

FIG. 13A provides a graph showing that SMN oligos 77 and 83 reduce EZH2 in SMN chromatin compared to controls (9677 cells only or lipid control). EZH2 ChIP data is shown.

FIG. 13B provides a graph showing H3K27m3 in SMN chromatin after treatment with SMN oligos 77 or 83 compared to controls (9677 cells only or lipid control). H3K27m3 ChIP data is shown.

FIG. 13C provides a graph showing the presence of H3, H3K4m3, H3K27m3, H3K36m3, RNA polII_S2 and EZH2 at the HOXC13 promoter after treatment withSMN oligos compared to controls (9677 cells only or lipid control).

FIG. 14A provides a graph showing H3K36m3 in SMN chromatin after treatment with SMN oligos 77 or 83 compared to controls (9677 cells only or lipid control). H3K36m3 ChIP data is shown.

FIG. 14B provides a graph showing RNA PolII S2 in SMN chromatin after treatment with SMN oligos 77 or 83 compared to controls (9677 cells only or lipid control). RNA PolII S2 ChIP data is shown.

FIG. 14C provides a graph showing H3K4m3 in SMN chromatin after treatment with SMN oligos 77 or 83 compared to controls (9677 cells only or lipid control). H3K4m3 ChIP data is shown.

FIG. 14D provides a graph showing Histone 3 (H3) in SMN chromatin after treatment with SMN oligos 77 or 83 compared to controls (9677 cells only or lipid control). H3 ChIP data is shown.

FIG. 15A provides a graph showing EZH2 in SMN chromatin after treatment with a splice switching oligo compared to controls (9677 cells only or lipid control). EZH2 ChIP data is shown.

FIG. 15B provides a graph showing H3K36m3 in SMN chromatin after treatment with a splice switching oligo compared to controls (9677 cells only or lipid control).

H3K36m3 ChIP data is shown. FIG. 15C provides a graph showing RNA PolII S2 in SMN chromatin after treatment with a splice switching oligo compared to controls (9677 cells only or lipid control). RNA PolII S2 ChIP data is shown.

FIG. 16A provides a graph showing the level of SMN1 mRNA in non-human primate (NHP) cells treated with various SMN oligos (37, 77, 83, 143, 144, or 243).

FIG. 16B provides a graph showing the level of SMN1 mRNA in non-human primate (NHP) cells treated with various splice- switiching SMN oligos (54m01 or 54ml5).

FIGs. 17A-D provides a series of graphs showing SMN2 mRNA levels in cells treated with an SMN oligo, a splice- switiching oligo, or a combination thereof. FIG. 17A shows data for a splice switching oligo at various concentrations alone or in combination with SMN oligo 143. FIG. 17B shows data for a splice switching oligo at various concentrations alone or in combination with SMN oligo 83. FIG. 17C shows data for SMN oligo 143 at various concentrations alone or in combination with a splice switching oligo. FIG. 17D shows data for SMN oligo 83 at various concentrations alone or in combination with a splice switching oligo.

FIG. 18 is a graph showing SMN2 mRNA increase in SMA patient fibroblast cell line GM09677 treated with various SMN oligos. Both SMN full length and exon 7 deleted (delta 7) mRNA levels are shown.

FIG. 19 is a graph showing SMN protein increase in SMA patient fibroblast cell line GM09677 treated with various SMN oligos.

FIG. 20 is a graph showing SMN2 mRNA increase in SMA patient fibroblast cell line GM09677 treated with various splice- switching oligos.

FIG. 21 is a graph showing SMN protein increase in SMA patient fibroblast cell line GM09677 treated with various splice- switching oligos.

BRIEF DESCRIPTION OF TABLES

Table 1: Hexamers that are not seed sequences of human miRNAs

Table 2: Oligonucleotide sequences made for testing in the lab. RQ (column 2) and RQ SE (column 3) shows the activity of the oligo relative to a control well (usually carrier alone) and the standard error or the triplicate replicates of the experiment, [oligo] is shown in nanomolar for in vitro experiments and in milligrams per kilogram of body weight for in vivo experiments. Table 3: A listing of oligonucleotide modifications. The suffix "Sup" in Table 3 indicates that a 3' end nucleotide may, for synthesis purposes, be conjugated to a solid support. It should be appreciated that in general when conjugated to a solid support for synthesis, the synthesized oligonucleotide is released such that the solid support is not part of the final oligonucleotide product.

Table 4: Oligonucleotide sequences made for testing . The table shows the sequence of the modified nucleotides, where InaX represents an LNA nucleotide with 3'

phosphorothioate linkage, omeX is a 2' -O-methyl nucleotide, dX is a deoxy nucleotide. An s at the end of a nucleotide code indicates that the nucleotide had a 3' phosphorothioate linkage. The "-Sup" at the end of the sequence marks the fact that the 3' end lacks either a phosphate or thiophosphate on the 3' linkage. The Formatted Sequence column shows the sequence of the oligonucleotide, including modified nucleotides, for the oligonucleotides tested in Table 2, 7, 8 and 9.

Table 5: Cell lines

Appendix A from International (PCT) Patent Application No.: PCT/US2013/041440, published as PCT Publication No.: WO/2013/173638, is incorporated herein by reference; Appendix A contains Table 7, which shows RT-PCR data from testing of different oligonucleotides.

Appendix B from International (PCT) Patent Application No.: PCT/US2013/041440, published as PCT Publication No.: WO/2013/173638, is incorporated herein by reference; Appendix B contains Table 8, which shows RT-PCR data from testing of different combination treatments (e.g. , two oligonucleotides, an oligonucleotide and a drug).

Appendix C from International (PCT) Patent Application No.: PCT/US2013/041440, published as PCT Publication No.: WO/2013/173638, is incorporated herein by reference; Appendix C contains Table 9, which shows ELISA data from testing of different

oligonucleotides

Note the following column information for Tables 7-9 in the above-referenced Appendices A-C, respectively. SEQID: sequence identifier of base sequence of

oligonucleotide used; Oligo Name: name of oligonucleotide; Avg RQ: average relative quantification of RT-PCR based expression levels of target gene(s); Avg RQ SE: standard error of mean of relative quantification of RT-PCR based expression level; " SMN over lipo only control" refers to the ratio of SMN protein levels (ng/mg total protein) when compared to Lipofectamine2000 (transfection reagent) treated cells converted into ; " SMN CVV" refers to coefficient of variation; Exp #: Experiment reference number; Target: target gene; [oligo]: concentration of oligonucleotide used in nM unless otherwise indicated; Cell Line: cell line used; Assay Type: assay used; Time(hr): time of assay following treatment; 2 nd Drug: name of second oligonucleotide (identified by Oligo Name) or drug used in

combination experiment; [2 nd ]: concentration of second oligonucleotide or drug; Units: units of concentration; 3 rd Drug: name of third oligonucleotide (identified by Oligo Name) or drug used in combination experiment; [3 rd ]: concentration of third oligonucleotide or drug; Notes: comments regarding experiment. Oligo Names correspond to those in Tables 2 and 4.

DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS OF THE INVENTION

Amyotrophic Lateral Sclerosis (ALS), also referred to as Lou Gehrig's disease and Charcot's disease, is a motor neuron disease characterized by rapidly progressive weakness, muscle atrophy and fasciculations, muscle spasticity, difficulty speaking (dysarthria), difficulty swallowing (dysphagia), and difficulty breathing (dyspnea), resulting in eventual death of the patient. These hallmarks of ALS are caused by damage to the neurons that control voluntary muscle movement.

Loss of Gems has been described in both ALS patient-derived cells and in animal models. It has been recognized that loss of Gems also occurs in Spinal Muscular Atrophy (SMA). Gems, or Gemini of coiled bodies, are compact structures that contain SMN protein and are found within cell nuclei. Gems are generally between 0.2 microns and 2 microns in diameter and, when viewed under an electron microscope, resemble a ball of tangled thread. Gems are thought to be involved in snRNP biogenesis. Mutant SOD1 alters SMN

localization and prevent Gem formation, which is restored by SMN over-expression. In a mutant SOD1 mouse model of ALS, overexpression of SMN delayed the loss of Gems and disease onset. Thus, in some embodiments, induction of SMN expression provides a therapeutic benefit in ALS (e.g., in familial ALS due to mutant SOD1 (-10% of ALS)). TDP-43 and FUS/TLS interact with SMN in nuclear Gems, and all three proteins function in spliceosome maintenance by controlling levels of U snRNAs. FUS is involved in Gem formation, and the loss of Gems in FUS-deficient cells can be reversed in cells over- expressing exogenous SMN. Thus, a common process involved in the pathogenesis of ALS may be the neuronal loss of spliceosome integrity resulting in abnormal splicing and motor neuron death, which may be due to abnormalities in SMN-containing Gems.

Accordingly, methods and related single stranded oligonucleotides are provided herein that are useful for selectively inducing expression of SMN in cells of a subject having a motor neuron disease, such as ALS, Primary Lateral Sclerosis, Progressive Muscular

Atrophy, Progressive Bulbar Palsy or Pseudobulbar Palsy. In some embodiments, methods are provided for inducing expression of particular splice variants of SMN1 or SMN2. Thus, in some embodiments, methods are provided that are useful for controlling the levels in a cell of particular SMN protein isoforms encoded by the splice variants. In some cases, the methods are useful for inducing expression of SMN proteins to levels sufficient to treat ALS. For example, according to some aspects of the invention methods are provided for increasing expression of full-length SMN protein in a cell for purposes of treating ALS. In some embodiments, the methods comprise delivering to the cell a first single stranded

oligonucleotide complementary with a PRC2-associated region of SMN1 or SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN1 or SMN2, in amounts sufficient to increase expression of a mature mRNA of SMN1 or SMN2 that comprises (or includes) exon 7 in the cell. Further aspects of the invention are described in detailed herein.

Accordingly, in some embodiments, methods provided herein comprise a step of administering a single- stranded oligonucleotide as provided herein to a cell or a subject comprising a mutation in a gene selected from SOD1, FUS/TLS, or TDP-43. Mutations in SOD1, FUS/TLS, and TDP-43 have been associated with ALS in humans and animal models. Mutations in SOD1 associated with ALS include A4V, H46R, G37R, L38V, and G93A.

Mutations in TDP-43 associated with ALS include D169G, K263E, N267S, G287S, G290A, S292N, G294A, G294V, G295S, G295R, G298S, M311V, A315T, A321G, A321V, Q331K, S332N, G335D, M337V, Q343R, N345K, G348C, G348V, N352S, N352T, R361S, P363A, Y374X, N378D, S379P, S379C, A382T, A382P, I383V, G384R, N390S, N390D, and S393L. Mutations in FUS TLS associated with ALS include S57del, G156E, G191S, R216C, G225V, G230C, R234C, R234L, R224C, M254V, S402_P441delinsGGGG, S462F, G466VfsX14, R495X, G507D, K510E, S513P, R514G, R514S, G515C, H517Q, H517P, R518K, R518G, R521H, R521G, R521C, R522G, R524S, R524T, and P525L. As described above, mutations in these genes have been shown to affect SMN-containing Gem formation and/or activity.

Mutations in other genes and loci in addition to SODl, FUS/TLS, and TDP-43 have also been associated with ALS. These genes are listed in the table below.

Accordingly, in some embodiments, methods provided herein comprise a step of administering a single- stranded oligonucleotide as provided herein to a cell or a subject comprising a mutation associated with ALS in a gene selected from SODl, ALS2, SETX, FUS/TLS, VAPB, ANG, TDP-43, FIG4, OPTN, ATXN2, VCP, UBQLN2, SIGMAR1, CHMP2B, PFNl,or C9orf72 with an oligonucleotide as provided herein.

In some embodiments, the cell is a cell obtained from or present in a subject having ALS. In some embodiments, the cell is a motor neuron. Motor neurons are efferent nerves (also called effector neurons) that carry signals from the spinal cord to the muscles to produce movement. A motor neuron may directly or indirectly carry signals to the muscles.

Amyotrophic Lateral Sclerosis (ALS) Aspects of the invention relate to subjects having ALS or cells or tissues obtained from subjects having ALS. ALS is a motor neuron disease characterized by degeneration of the upper and lower motor neurons, which eventually leads to muscle weakness and atrophy throughout the body.

In some embodiments, a subject treated with an oligonucleotide disclosed herein is a subject having one or more symptoms of ALS. ALS may be diagnosed based on symptoms (which may be identified through a physical examination by a medical professional) and/or a series of tests, some of which are designed to distinguish ALS from other diseases that have similar symptoms to ALS. Symptoms of ALS include difficulty breathing, difficulty swallowing (e.g., choking easily, drooling, or gagging), head drop due to weakness of the neck muscles, muscle cramps, muscle contractions called fasciculations, muscle weakness that slowly gets worse (commonly involves one part of the body first, such as the arm or hand, and may eventually lead to difficulty lifting, climbing stairs, and walking), paralysis, speech problems (e.g., slow or abnormal speech pattern), voice changes, hoarseness, and weight loss. A physical examination may be used to identify such symptoms in a patient by examining the patient for one or more of the following: weakness, muscle tremors, muscle spasms, muscle twitching, loss of muscle tissue, twitching of the tongue, abnormal reflexes, stiff or clumsy walk, increased reflexes at the joints, difficulty controlling crying or laughing (sometimes called emotional incontinence), or loss of gag reflex. Exemplary tests that may be used to diagnose or aid in diagnosis of ALS include, but are not limited to: blood and/or urine tests to distinguish ALS from other conditions; breathing test to see if lung muscles are affected; cervical spine CT or MRI to be identify disease or injury to the neck, which can mimic ALS; electromyography to see which nerves or muscles do not work properly; nerve conduction study; genetic testing; head CT or MRI to distinguish ALS from other conditions; swallowing studies; and spinal tap (lumbar puncture). Other conditions or diseases that cause ALS-like symptoms include infectious diseases (such as human immunodeficiency virus (HIV), human T-cell leukemia virus (HTLV), polio, West Nile virus, and Lyme disease), multiple sclerosis, post-polio syndrome, multifocal motor neuropathy, and spinal muscular atrophy.

In some embodiments, a subject diagnosed with ALS may be monitored for progression of the disease. In some embodiments, the subject is monitored using the ALS functional rating scale (ALSFRS, see, e.g., The Amyotrophic Lateral Sclerosis Functional Rating Scale: Assessment of Activities of Daily Living in Patients With Amyotrophic Lateral Sclerosis. Arch Neurol. 1996;53(2): 141-147) or the revised ALS functional rating scale (ALSFRS-R, see, e.g., Cedarbaum JM, Stambler N, Malta E, Fuller C, Hilt D, Thurmond B, Nakanishi A) or another appropriate scale. The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. BDNF ALS Study Group (Phase III). J Neurol Sci. 1999 Oct 31;169(l-2): 13-21). The ALSFRS and ALSFRS-R measure the following categories of behavior: speech, salivation, swallowing, handwriting, cutting food and handling utensils (with or without gastrostomy), dressing and hygiene, turning in bed and adjusting bed clothes, walking, climbing stairs, and breathing. The ALSFRS-R further includes additional assessments of dyspnea, orthopnea, and the need for ventilatory support. An exemplary ALSFRS-R test is provided through the Center for Outcomes Research website under the ALS C.A.R.E project (see ALS Functional Rating Scale Scoring Tool Online). In some embodiments, a score may be calculated based on the characteristic of each of these categories (normal or slight disturbance giving a high score for each category, and marked or severe disturbance giving a low score for each category). In some

embodiments, the score for each category is then added together to create a total score. In some embodiments, the test may be given multiple times, in order to track changes in the total score over time. In some embodiments, an increase may indicate progression of ALS; whereas decrease or unchanging score may indicate stabilization or regression of ALS.

Poly comb repressive complex 2 (PRC2 )-interacting RNAs

Aspects of the invention provided herein relate to the discovery of polycomb repressive complex 2 (PRC2)-interacting RNAs. Polycomb repressive complex 2 (PRC2) is a histone methyltransferase and a known epigenetic regulator involved in silencing of genomic regions through methylation of histone H3. Among other functions, PRC2 interacts with long noncoding RNAs (IncRNAs), such as Rep A, Xist, and Tsix, to catalyze

trimethylation of histone H3-lysine27. PRC2 contains four subunits, Eed, Suzl2, RbAp48, and Ezh2. Aspects of the invention relate to the recognition that single stranded

oligonucleotides that bind to PRC2-associated regions of RNAs (e.g., IncRNAs) that are expressed from within a genomic region that encompasses or that is in functional proximity to the SMN1 or SMN2 gene can induce or enhance expression of SMN1 or SMN2. In some embodiments, this upregulation is believed to result from inhibition of PRC2 mediated repression of SMN1 or SMN2.

As used herein, the term "PRC2-associated region" refers to a region of a nucleic acid that comprises or encodes a sequence of nucleotides that interact directly or indirectly with a component of PRC2. A PRC2-associated region may be present in a RNA (e.g. , a long non- coding RNA (IncRNA)) that interacts with a PRC2. A PRC2-associated region may be present in a DNA that encodes an RNA that interacts with PRC2. In some cases, the PRC2- associated region is equivalently referred to as a PRC2-interacting region.

In some embodiments, a PRC2-associated region is a region of an RNA that crosslinks to a component of PRC2 in response to in situ ultraviolet irradiation of a cell that expresses the RNA, or a region of genomic DNA that encodes that RNA region. In some embodiments, a PRC2-associated region is a region of an RNA that immunoprecipitates with an antibody that targets a component of PRC2, or a region of genomic DNA that encodes that RNA region. In some embodiments, a PRC2-associated region is a region of an RNA that immunoprecipitates with an antibody that binds specifically to SUZ12, EED, EZH2 or

RBBP4 (which as noted above are components of PRC2), or a region of genomic DNA that encodes that RNA region.

In some embodiments, a PRC2-associated region is a region of an RNA that is protected from nucleases (e.g. , RNases) in an RNA-immunoprecipitation assay that employs an antibody that targets a component of PRC2, or a region of genomic DNA that encodes that protected RNA region. In some embodiments, a PRC2-associated region is a region of an RNA that is protected from nucleases (e.g. , RNases) in an RNA-immunoprecipitation assay that employs an antibody that targets SUZ12, EED, EZH2 or RBBP4, or a region of genomic DNA that encodes that protected RNA region.

In some embodiments, a PRC2-associated region is a region of an RNA within which occur a relatively high frequency of sequence reads in a sequencing reaction of products of an RNA-immunoprecipitation assay that employs an antibody that targets a component of PRC2, or a region of genomic DNA that encodes that RNA region. In some embodiments, a PRC2- associated region is a region of an RNA within which occur a relatively high frequency of sequence reads in a sequencing reaction of products of an RNA-immunoprecipitation assay that employs an antibody that binds specifically to SUZ12, EED, EZH2 or RBBP4, or a region of genomic DNA that encodes that protected RNA region. In such embodiments, the PRC2-associated region may be referred to as a "peak."

In some embodiments, a PRC2-associated region comprises a sequence of 40 to 60 nucleotides that interact with PRC2 complex. In some embodiments, a PRC2-associated region comprises a sequence of 40 to 60 nucleotides that encode an RNA that interacts with PRC2. In some embodiments, a PRC2-associated region comprises a sequence of up to 5kb in length that comprises a sequence (e.g. , of 40 to 60 nucleotides) that interacts with

PRC2. In some embodiments, a PRC2-associated region comprises a sequence of up to 5kb in length within which an RNA is encoded that has a sequence (e.g. , of 40 to 60 nucleotides) that is known to interact with PRC2. In some embodiments, a PRC2-associated region comprises a sequence of about 4kb in length that comprise a sequence (e.g. , of 40 to 60 nucleotides) that interacts with PRC2. In some embodiments, a PRC2-associated region comprises a sequence of about 4kb in length within which an RNA is encoded that includes a sequence (e.g. , of 40 to 60 nucleotides) that is known to interact with PRC2. In some embodiments, a PRC2-associated region has a sequence as set forth in any one of SEQ ID

NOS: 9 to 29. In some embodiments, a PRC2-associated region has a sequence as set forth in any one of SEQ ID NOS: 24 to 29.

In some embodiments, single stranded oligonucleotides are provided that specifically bind to, or are complementary to, a PRC2-associated region in a genomic region that encompasses or that is in proximity to the SMN1 or SMN2 gene. In some embodiments, single stranded oligonucleotides are provided that specifically bind to, or are complementary to, a PRC2-associated region that has a sequence as set forth in any one of SEQ ID NOS: 9 to 29. In some embodiments, single stranded oligonucleotides are provided that specifically bind to, or are complementary to, a PRC2-associated region that has a sequence as set forth in any one of SEQ ID NOS: 9 to 29 combined with up to 2kb, up to 5kb, or up to lOkb of flanking sequences from a corresponding genomic region to which these SEQ IDs map (e.g. , in a human genome). In some embodiments, single stranded oligonucleotides have a sequence as set forth in any one of SEQ ID NOS: 30 to 13087 or 13108 to 13116. In some embodiments, single stranded oligonucleotides have a sequence as set forth in Table 2. In some embodiments, a PRC2 associated region of SMN1 or SMN2 against which a single stranded oligonucleotide is complementary is selected from SEQ ID NOS: 24-29. In some embodiments, a single stranded oligonucleotide that is complementary with a PRC2 associated region of SMNl or SMN2 comprises a sequence selected from SEQ ID NOS: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, and 13108-13116. In some embodiments, a single stranded oligonucleotide that is complementary with a PRC2 associated region of SMNl or SMN2 comprises a sequence selected from 11395, 11394, 10169, and 10170.

Without being bound by a theory of invention, these oligonucleotides are able to interfere with the binding of and function of PRC2, by preventing recruitment of PRC2 to a specific chromosomal locus. For example, a single administration of single stranded oligonucleotides designed to specifically bind a PRC2-associated region IncRNA can stably displace not only the IncRNA, but also the PRC2 that binds to the IncRNA, from binding chromatin. After displacement, the full complement of PRC2 is not recovered for up to 24 hours. Further, IncRNA can recruit PRC2 in a cis fashion, repressing gene expression at or near the specific chromosomal locus from which the IncRNA was transcribed.

Methods of modulating gene expression are provided, in some embodiments, that may be carried out in vitro, ex vivo, or in vivo. It is understood that any reference to uses of compounds throughout the description contemplates use of the compound in preparation of a pharmaceutical composition or medicament for use in the treatment of condition (e.g., ALS) associated with decreased levels or activity of SMNl or SMN2. Thus, as one nonlimiting example, this aspect of the invention includes use of such single stranded oligonucleotides in the preparation of a medicament for use in the treatment of disease, wherein the treatment involves upregulating expression of SMNl or SMN2.

In further aspects of the invention, methods are provided for selecting a candidate oligonucleotide for activating expression of SMNl or SMN2. The methods generally involve selecting as a candidate oligonucleotide, a single stranded oligonucleotide comprising a nucleotide sequence that is complementary to a PRC2-associated region (e.g., a nucleotide sequence as set forth in any one of SEQ ID NOS: 9 to 29). In some embodiments, sets of oligonucleotides may be selected that are enriched (e.g., compared with a random selection of oligonucleotides) in oligonucleotides that activate expression of SMNl or SMN2. Single Stranded Oligonucleotides for Modulating Expression of SMNl or SMN2 In one aspect of the invention, single stranded oligonucleotides complementary to the PRC2-associated regions are provided for modulating expression of SMNl or SMN2 in a cell. In some embodiments, expression of SMNl or SMN2 is upregulated or increased. In some embodiments, single stranded oligonucleotides complementary to these PRC2- associated regions inhibit the interaction of PRC2 with long RNA transcripts such that gene expression is upregulated or increased. In some embodiments, single stranded

oligonucleotides complementary to these PRC2- associated regions inhibit the interaction of PRC2 with long RNA transcripts, resulting in reduced methylation of histone H3 and reduced gene inactivation, such that gene expression is upregulated or increased. In some

embodiments, this interaction may be disrupted or inhibited due to a change in the structure of the long RNA that prevents or reduces binding to PRC2. The oligonucleotide may be selected using any of the methods disclosed herein for selecting a candidate oligonucleotide for activating expression of SMNl or SMN2.

The single stranded oligonucleotide may comprise a region of complementarity that is complementary with a PRC2-associated region of a nucleotide sequence set forth in any one of SEQ ID NOS: 1 to 8. The region of complementarity of the single stranded

oligonucleotide may be complementary with at least 6, e.g., at least 7, at least 8, at least 9, at least 10, at least 15 or more consecutive nucleotides of the PRC2-associated region.

It should be appreciated that due the high homology between SMNl and SMN2, single stranded oligonucleotides that are complementary with a PRC2-associated region of SMNl are often also complementary with a corresponding PRC2-associated region of SMN2.

The PRC2-associated region may map to a position in a chromosome between 50 kilobases upstream of a 5 '-end of the SMNl or SMN2 gene and 50 kilobases downstream of a 3 '-end of the SMNl or SMN2 gene. The PRC2-associated region may map to a position in a chromosome between 25 kilobases upstream of a 5 '-end of the SMNl or SMN2 gene and 25 kilobases downstream of a 3 '-end of the SMNl or SMN2 gene. The PRC2-associated region may map to a position in a chromosome between 12 kilobases upstream of a 5'-end of the SMNl or SMN2 gene and 12 kilobases downstream of a 3 '-end of the SMNl or SMN2 gene. The PRC2-associated region may map to a position in a chromosome between 5 kilobases upstream of a 5 '-end of the SMNl or SMN2 gene and 5 kilobases downstream of a 3 '-end of the SMNl or SMN2 gene. The genomic position of the selected PRC2-associated region relative to the SMNl or SMN2 gene may vary. For example, the PRC2-associated region may be upstream of the 5' end of the SMNl or SMN2 gene. The PRC2-associated region may be downstream of the 3' end of the SMNl or SMN2 gene. The PRC2-associated region may be within an intron of the SMNl or SMN2 gene. The PRC2-associated region may be within an exon of the SMNl or SMN2 gene. The PRC2-associated region may traverse an intron-exon junction, a 5'-UTR- exon junction or a 3'-UTR-exon junction of the SMNl or SMN2 gene.

The single stranded oligonucleotide may comprise a sequence having the formula X- Y-Z, in which X is any nucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a human seed sequence of a microRNA, and Z is a nucleotide sequence of varying length. In some embodiments X is the 5' nucleotide of the oligonucleotide. In some embodiments, when X is anchored at the 5' end of the oligonucleotide, the oligonucleotide does not have any nucleotides or nucleotide analogs linked 5' to X. In some embodiments, other compounds such as peptides or sterols may be linked at the 5' end in this embodiment as long as they are not nucleotides or nucleotide analogs. In some embodiments, the single stranded oligonucleotide has a sequence 5'X-Y-Z and is 8-50 nucleotides in length.

Oligonucleotides that have these sequence characteristics are predicted to avoid the miRNA pathway. Therefore, in some embodiments, oligonucleotides having these sequence characteristics are unlikely to have an unintended consequence of functioning in a cell as a miRNA molecule. The Y sequence may be a nucleotide sequence of 6 nucleotides in length set forth in Table 1.

The single stranded oligonucleotide may have a sequence that does not contain guanosine nucleotide stretches (e.g. , 3 or more, 4 or more, 5 or more, 6 or more consecutive guanosine nucleotides). In some embodiments, oligonucleotides having guanosine nucleotide stretches have increased non-specific binding and/or off-target effects, compared with oligonucleotides that do not have guanosine nucleotide stretches.

The single stranded oligonucleotide may have a sequence that has less than a threshold level of sequence identity with every sequence of nucleotides, of equivalent length, that map to a genomic position encompassing or in proximity to an off-target gene. For example, an oligonucleotide may be designed to ensure that it does not have a sequence that maps to genomic positions encompassing or in proximity with all known genes (e.g. , all known protein coding genes) other than SMNl or SMN2. In a similar embodiment, an oligonucleotide may be designed to ensure that it does not have a sequence that maps to any other known PRC2-associated region, particularly PRC2-associated regions that are functionally related to any other known gene (e.g. , any other known protein coding gene). In either case, the oligonucleotide is expected to have a reduced likelihood of having off-target effects. The threshold level of sequence identity may be 50%, 60%, 70%, 80%, 85%, 90%, 95%, 99% or 100% sequence identity.

The single stranded oligonucleotide may have a sequence that is complementary to a PRC2-associated region that encodes an RNA that forms a secondary structure comprising at least two single stranded loops. In has been discovered that, in some embodiments, oligonucleotides that are complementary to a PRC2-associated region that encodes an RNA that forms a secondary structure comprising one or more single stranded loops (e.g. , at least two single stranded loops) have a greater likelihood of being active (e.g. , of being capable of activating or enhancing expression of a target gene) than a randomly selected

oligonucleotide. In some cases, the secondary structure may comprise a double stranded stem between the at least two single stranded loops. Accordingly, the region of

complementarity between the oligonucleotide and the PRC2-associated region may be at a location of the PRC2 associated region that encodes at least a portion of at least one of the loops. In some cases, the region of complementarity between the oligonucleotide and the PRC2-associated region may be at a location of the PRC2-associated region that encodes at least a portion of at least two of the loops. In some cases, the region of complementarity between the oligonucleotide and the PRC2-associated region may be at a location of the PRC2 associated region that encodes at least a portion of the double stranded stem. In some embodiments, a PRC2-associated region (e.g. , of an IncRNA) is identified (e.g. , using RIP- Seq methodology or information derived therefrom). In some embodiments, the predicted secondary structure RNA (e.g. , IncRNA) containing the PRC2-associated region is determined using RNA secondary structure prediction algorithms, e.g. , RNAfold, mfold. In some embodiments, oligonucleotides are designed to target a region of the RNA that forms a secondary structure comprising one or more single stranded loop (e.g. , at least two single stranded loops) structures which may comprise a double stranded stem between the at least two single stranded loops.

The single stranded oligonucleotide may have a sequence that is has greater than 30% G-C content, greater than 40% G-C content, greater than 50% G-C content, greater than 60% G-C content, greater than 70% G-C content, or greater than 80% G-C content. The single stranded oligonucleotide may have a sequence that has up to 100% G-C content, up to 95% G-C content, up to 90% G-C content, or up to 80% G-C content. In some embodiments in which the oligonucleotide is 8 to 10 nucleotides in length, all but 1, 2, 3, 4, or 5 of the nucleotides of the complementary sequence of the PRC2-associated region are cytosine or guanosine nucleotides. In some embodiments, the sequence of the PRC2- associated region to which the single stranded oligonucleotide is complementary comprises no more than 3 nucleotides selected from adenine and uracil.

The single stranded oligonucleotide may be complementary to a chromosome of a different species (e.g., a mouse, rat, rabbit, goat, monkey, etc.) at a position that encompasses or that is in proximity to that species' homolog of SMN1 or SMN2. The single stranded oligonucleotide may be complementary to a human genomic region encompassing or in proximity to the SMN1 or SMN2 gene and also be complementary to a mouse genomic region encompassing or in proximity to the mouse homolog of SMN1 or SMN2. For example, the single stranded oligonucleotide may be complementary to a sequence as set forth in SEQ ID NO: 1, 2, 4, or 5, which is a human genomic region encompassing or in proximity to the SMN1 or SMN2 gene, and also be complementary to a sequence as set forth in SEQ ID NO:7 or 8, which is a mouse genomic region encompassing or in proximity to the mouse homolog of the SMN1 or SMN2 gene. Oligonucleotides having these characteristics may be tested in vivo or in vitro for efficacy in multiple species (e.g., human and mouse). This approach also facilitates development of clinical candidates for treating human disease by selecting a species in which an appropriate animal exists for the disease.

In some embodiments, the region of complementarity of the single stranded oligonucleotide is complementary with at least 8 to 15, 8 to 30, 8 to 40, or 10 to 50, or 5 to 50, or 5 to 40 bases, e.g., 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 consecutive nucleotides of a PRC2-associated region. In some embodiments, the region of complementarity is complementary with at least 8 consecutive nucleotides of a PRC2-associated region. In some embodiments the sequence of the single stranded oligonucleotide is based on an RNA sequence that binds to PRC2, or a portion thereof, said portion having a length of from 5 to 40 contiguous base pairs, or about 8 to 40 bases, or about 5 to 15, or about 5 to 30, or about 5 to 40 bases, or about 5 to 50 bases. Complementary, as the term is used in the art, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of PRC2-associated region, then the single stranded nucleotide and PRC2-associated region are considered to be complementary to each other at that position. The single stranded nucleotide and PRC2-associated region are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides that can hydrogen bond with each other through their bases. Thus, "complementary" is a term which is used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the single stranded nucleotide and PRC2-associated region. For example, if a base at one position of a single stranded nucleotide is capable of hydrogen bonding with a base at the corresponding position of a PRC2-associated region, then the bases are considered to be complementary to each other at that position. 100% complementarity is not required.

The single stranded oligonucleotide may be at least 80% complementary to

(optionally one of at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% complementary to) the consecutive nucleotides of a PRC2-associated region. In some embodiments the single stranded oligonucleotide may contain 1, 2 or 3 base mismatches compared to the portion of the consecutive nucleotides of a PRC2-associated region. In some embodiments the single stranded oligonucleotide may have up to 3 mismatches over 15 bases, or up to 2 mismatches over 10 bases.

It is understood in the art that a complementary nucleotide sequence need not be 100% complementary to that of its target to be specifically hybridizable. In some

embodiments, a complementary nucleic acid sequence for purposes of the present disclosure is specifically hybridizable when binding of the sequence to the target molecule (e.g., IncRNA) interferes with the normal function of the target (e.g., IncRNA) to cause a loss of activity (e.g., inhibiting PRC2-associated repression with consequent up-regulation of gene expression) and there is a sufficient degree of complementarity to avoid non-specific binding of the sequence to non-target sequences under conditions in which avoidance of non-specific binding is desired, e.g., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed under suitable conditions of stringency. In some embodiments, the single stranded oligonucleotide is 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50 or more nucleotides in length. In a preferred embodiment, the oligonucleotide is 8 to 30 nucleotides in length.

In some embodiments, the PRC2-associated region occurs on the same DNA strand as a gene sequence (sense). In some embodiments, the PRC2-associated region occurs on the opposite DNA strand as a gene sequence (anti-sense). Oligonucleotides complementary to a PRC2-associated region can bind either sense or anti-sense sequences. Base pairings may include both canonical Watson-Crick base pairing and non-Watson-Crick base pairing (e.g. , Wobble base pairing and Hoogsteen base pairing). It is understood that for complementary base pairings, adenosine-type bases (A) are complementary to thymidine-type bases (T) or uracil-type bases (U), that cytosine-type bases (C) are complementary to guanosine-type bases (G), and that universal bases such as 3-nitropyrrole or 5-nitroindole can hybridize to and are considered complementary to any A, C, U, or T. Inosine (I) has also been considered in the art to be a universal base and is considered complementary to any A, C, U or T.

In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) or uridine (U) nucleotides (or a modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be replaced with any other nucleotide suitable for base pairing (e.g., via a Watson-Crick base pair) with an adenosine nucleotide. In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) or uridine (U) nucleotides (or a modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be suitably replaced with a different pyrimidine nucleotide or vice versa. In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be suitably replaced with a uridine (U) nucleotide (or a modified nucleotide thereof) or vice versa. In some embodiments, GC content of the single stranded oligonucleotide is preferably between about 30-60 %. Contiguous runs of three or more Gs or Cs may not be preferable in some embodiments. Accordingly, in some embodiments, the oligonucleotide does not comprise a stretch of three or more guanosine nucleotides.

In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to an RNA that is encoded in a genome (e.g., a human genome) as a single contiguous transcript (e.g., a non-spliced RNA). In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to an RNA that is encoded in a genome (e.g., a human genome), in which the distance in the genome between the 5 'end of the coding region of the RNA and the 3' end of the coding region of the RNA is less than 1 kb, less than 2 kb, less than 3 kb, less than 4 kb, less than 5 kb, less than 7 kb, less than 8 kb, less than 9 kb, less than 10 kb, or less than 20 kb.

It is to be understood that any oligonucleotide provided herein can be excluded.

In some embodiments, single stranded oligonucleotides disclosed herein may increase expression of mRNA corresponding to the gene by at least about 50% (i.e. 150% of normal or 1.5 fold), or by about 2 fold to about 5 fold. In some embodiments, expression may be increased by at least about 15 fold, 20 fold, 30 fold, 40 fold, 50 fold or 100 fold, or any range between any of the foregoing numbers. It has also been found that increased mRNA expression has been shown to correlate to increased protein expression.

In some or any of the embodiments of the oligonucleotides described herein, or processes for designing or synthesizing them, the oligonucleotides will upregulate gene expression and may specifically bind or specifically hybridize or be complementary to the PRC2 binding RNA that is transcribed from the same strand as a protein coding reference gene. The oligonucleotide may bind to a region of the PRC2 binding RNA that originates within or overlaps an intron, exon, intron exon junction, 5' UTR, 3' UTR, a translation initiation region, or a translation termination region of a protein coding sense strand of a reference gene (refGene).

In some or any of the embodiments of oligonucleotides described herein, or processes for designing or synthesizing them, the oligonucleotides will upregulate gene expression and may specifically bind or specifically hybridize or be complementary to a PRC2 binding RNA that transcribed from the opposite strand (the antisense strand) of a protein coding reference gene. The oligonucleotide may bind to a region of the PRC2 binding RNA that originates within or overlaps an intron, exon, intron exon junction, 5' UTR, 3' UTR, a translation initiation region, or a translation termination region of a protein coding antisense strand of a reference gene.

The oligonucleotides described herein may be modified, e.g., comprise a modified sugar moiety, a modified internucleoside linkage, a modified nucleotide and/or combinations thereof. In addition, the oligonucleotides can exhibit one or more of the following properties: do not induce substantial cleavage or degradation of the target RNA; do not cause

substantially complete cleavage or degradation of the target RNA; do not activate the RNAse H pathway; do not activate RISC; do not recruit any Argonaute family protein; are not cleaved by Dicer; do not mediate alternative splicing; are not immune stimulatory; are nuclease resistant; have improved cell uptake compared to unmodified oligonucleotides; are not toxic to cells or mammals; may have improved endosomal exit; do interfere with interaction of IncRNA with PRC2, preferably the Ezh2 subunit but optionally the Suzl2, Eed, RbAp46/48 subunits or accessory factors such as Jarid2; do decrease histone H3 lysine27 methylation and/or do upregulate gene expression.

Oligonucleotides that are designed to interact with RNA to modulate gene expression are a distinct subset of base sequences from those that are designed to bind a DNA target (e.g., are complementary to the underlying genomic DNA sequence from which the RNA is transcribed). Splice Switching Oligonucleotides

Aspects of the invention provide strategies for targeting SMN1 or SMN2 precursor mRNA to affect splicing to minimize exon skipping. Accordingly, aspects of the invention provide therapeutic compounds useful for the treatment of ALS. In some embodiments, oligonucleotides, referred to herein as "splice switching oligonucleotides" are provided that modulate SMN2 splicing. Methods and related compositions, compounds, and kits are provided, in some embodiments, that are useful for increasing expression of full-length.

SMN protein in a cell. The methods generally involve delivering to a cell a first single stranded oligonucleotide complementary with at least 8 consecutive nucleotides of a PRC2- associated region of SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMN2, in amounts sufficient to increase expression of a mature mRNA of SMN2 that comprises (or includes) exon 7 in the cell. Any of the single stranded oligonucleotides that are complementary with at least 8 consecutive nucleotides of a PRC2- associated region of SMN1 or SMN2 may be used. It should be appreciated that single stranded oligonucleotides that are complementary with a splice control sequence may alternatively be referred herein, as splice switching oligonucleotides. Splice switching oligonucleotides typically comprise a sequence complementary to a splice control sequence (e.g. , a intronic splicing silencer sequence) of a precursor mRNA, and are capable of binding to and affecting processing of the precursor mRNA. Splice switching oligonucleotides may be complementary with a region of an exon, a region of an intron or an intron/exon junction. In some embodiments, the splice control sequence comprises the sequence: CAGCAUUAUGAAAG (SEQ ID NO: 13100) or a portion thereof. In some embodiments, the splice control sequence comprises at least one hnRNAP binding sequence. In some embodiments, splice switching oligonucleotides that target SMN1 or SMN2 function based on the premise that there is a competition between the 3' splice sites of exons 7 and 8 for pairing with the 5' splice site of exon 6, so impairing the recognition of the 3' splice site of exon 8 favors exon 7 inclusion. In some embodiments, splice switching oligonucleotides are provided that promote SMN2 exon 7 inclusion and full-length SMN protein expression, in which the oligonucleotides are complementary to the intron 7/exon 8 junction. In some embodiments, splice switching oligonucleotide are composed of a segment complementary to an exon of SMN1 or SMN2 (e.g., exon 7). In some embodiments, splice switching oligonucleotides comprise a tail (e.g. , a non-complementary tail) consisting of RNA sequences with binding motifs recognized by a serine/arginine-rich (SR) protein. In some embodiments, splice switching oligonucleotides are complementary (at least partially) with an intronic splicing silencer (ISS). In some embodiments, the ISS is in intron 6 or intron 7 of SMN1 or SMN2. In some embodiments, splice switching oligonucleotides comprise an antisense moiety complementary to a target exon or intron (e.g., of SMN1 or SMN2) and a minimal RS domain peptide similar to the splicing activation domain of SR proteins. In some embodiments, the splice switching oligonucleotide is 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50 or more nucleotides in length. In one embodiment, the oligonucleotide is 8 to 30 nucleotides in length.

Linkers

Any of the oligonucleotides disclosed herein may be linked to one or more other oligonucleotides disclosed herein by a linker, e.g. , a cleavable linker. Accordingly, in some embodiments, compounds are provided that comprise a single stranded oligonucleotide complementary with a PRC2-associated region of a gene that is linked via a linker to a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, compounds are provided that have the general formula A-B-C, in which A is a single stranded oligonucleotide complementary with a PRC2- associated region of a gene, B is a linker, and C is a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, linker B comprises an oligonucleotide, peptide, low pH labile bond, or disulfide bond. In some embodiments, the compounds is orientated as 5'-A-B-C-3'. In some embodiments, the compound is orientated as 3' -A-B-C-5' . In some embodiments, where B is an oligonucleotide, the 3' end of A is linked to the 5' end of B, and the 3' end of B is linked to 5' end of C. In some embodiments, where B is an oligonucleotide, the 5' end of A is linked to the 3' end of B, and the 5' end of B is linked to 3' end of C. In some

embodiments, where B is an oligonucleotide, the 5' end of A is linked to the 5' end of B, and/or the 3' end of B is linked to the 3' end of C. In some embodiments, where B is an oligonucleotide, the 3' end of A is linked to the 3' end of B, and/or the 5' end of B is linked to the 5' end of C.

The term "linker" generally refers to a chemical moiety that is capable of covalently linking two or more oligonucleotides. In some embodiments, at least one bond comprised or contained within the linker is capable of being cleaved (e.g., in a biological context, such as in a mammalian extract, such as an endosomal extract), such that at least two

oligonucleotides are no longer covalently linked to one another after bond cleavage. It will be appreciated that, in some embodiments, a provided linker may include a region that is non- cleavable, as long as the linker also comprises at least one bond that is cleavable.

In some embodiments, the linker comprises a polypeptide that is more susceptible to cleavage by an endopeptidase in the mammalian extract than the oligonucleotides. The endopeptidase may be a trypsin, chymotrypsin, elastase, thermolysin, pepsin, or

endopeptidase V8. The endopeptidase may be a cathepsin B, cathepsin D, cathepsin L, cathepsin C, papain, cathepsin S or endosomal acidic insulinase. For example, the linker comprise a peptide having an amino acid sequence selected from: ALAL, APISFFELG, FL, GFN, R/KXX, GRWHTVGLRWE, YL, GF, and FF, in which X is any amino acid.

In some embodiments, the linker comprises the formula -(CH2),jS-S(CH2)m-, wherein n and m are independently integers from 0 to 10.

In some embodiments, the linker may comprise an oligonucleotide that is more susceptible to cleavage by an endonuclease in the mammalian extract than the oligonucleotides. The linker may have a nucleotide sequence comprising from 1 to 10 thymidines or uridines. The linker may have a nucleotide sequence comprising

deoxyribonucleotides linked through phosphodiester intemucleotide linkages. The linker may have a nucleotide sequence comprising from 1 to 10 thymidines or uridines linked through phosphodiester intemucleotide linkages. The linker may have a nucleotide sequence comprising from 1 to 10 thymidines or uridines linked through phosphorothioate

intemucleotide linkages.

In some embodiments, at least one linker is 2-fold, 3-fold, 4-fold, 5-fold, 10-fold or more sensitive to enzymatic cleavage in the presence of a mammalian extract than at least two oligonucleotides. It should be appreciated that different linkers can be designed to be cleaved at different rates and/or by different enzymes in compounds comprising two or more linkers. Similarly different linkers can be designed to be sensitive to cleavage in different tissues, cells or subcellular compartments in compounds comprising two or more linkers. This can advantageously permit compounds to have oligonucleotides that are released from compounds at different rates, by different enzymes, or in different tissues, cells or subcellular compartments thereby controlling release of the monomeric oligonucleotides to a desired in vivo location or at a desired time following administration.

In certain embodiments, linkers are stable (e.g., more stable than the oligonucleotides they link together) in plasma, blood or serum which are richer in exonucleases, and less stable in the intracellular environments which are relatively rich in endonucleases. In some embodiments, a linker is considered "non-cleavable" if the linker's half-life is at least 24, or 28, 32, 36, 48, 72, 96 hours or longer under the conditions described here, such as in liver homogenates. Conversely, in some embodiments, a linker is considered "cleavable" if the half-life of the linker is at most 10, or 8, 6, 5 hours or shorter.

In some embodiments, the linker is a nuclease-cleavable oligonucleotide linker. In some embodiments, the nuclease-cleavable linker contains one or more phosphodiester bonds in the oligonucleotide backbone. For example, the linker may contain a single

phosphodiester bridge or 2, 3, 4, 5, 6, 7 or more phosphodiester linkages, for example as a string of 1-10 deoxynucleotides, e.g., dT, or ribonucleotides, e.g., rU, in the case of RNA linkers. In the case of dT or other DNA nucleotides dN in the linker, in certain embodiments the cleavable linker contains one or more phosphodiester linkages. In other embodiments, in the case of rU or other RNA nucleotides rN, the cleavable linker may consist of phosphorothioate linkages only. In contrast to phosphorothioate-linked deoxynucleotides, which in some embodiments are cleaved relatively slowly by nucleases (thus termed

"noncleavable"), phosphorothioate-linked rU undergoes relatively rapid cleavage by ribonucleases and therefore is considered cleavable herein in some embodiments. It is also possible to combine dN and rN into the linker region, which are connected by phosphodiester or phosphorothioate linkages. In other embodiments, the linker can also contain chemically modified nucleotides, which are still cleavable by nucleases, such as, e.g., 2'-0-modified analogs. In particular, 2'-0-methyl or 2'-fluoro nucleotides can be combined with each other or with dN or rN nucleotides. Generally, in the case of nucleotide linkers, the linker is a part of the compound that is usually not complementary to a target, although it could be. This is because the linker is generally cleaved prior to action of the oligonucleotides on the target, and therefore, the linker identity with respect to a target is inconsequential. Accordingly, in some embodiments, a linker is an (oligo)nucleotide linker that is not complementary to any of the targets against which the oligonucleotides are designed.

In some embodiments, the cleavable linker is an oligonucleotide linker that contains a continuous stretch of deliberately introduced Rp phosphorothioate stereoisomers (e.g., 4, 5, 6, 7 or longer stretches). The Rp stereoisoform, unlike Sp isoform, is known to be susceptible to nuclease cleavage (Krieg et al., 2003, Oligonucleotides, 13:491-499). Such a linker would not include a racemic mix of PS linkaged oligonucleotides since the mixed linkages are relatively stable and are not likely to contain long stretches of the Rp stereoisomers, and therefore, considered "non-cleavable" herein. Thus, in some embodiments, a linker comprises a stretch of 4, 5, 6, 7 or more phosphorothioated nucleotides, wherein the stretch does not contain a substantial amount or any of the Sp stereoisoform. The amount could be considered substantial if it exceeds 10% on a per- mole basis.

In some embodiments, the linker is a non-nucleotide linker, for example, a single phosphodiester bridge. Another example of such cleavable linkers is a chemical group comprising a disulfide bond, for example, -(CH 2 )„S-S(CH 2 ) m -, wherein n and m are integers from 0 to 10. In illustrative embodiments, n=m=6. Additional examples of non-nucleotide linkers are described below.

The linker can be designed so as to undergo a chemical or enzymatic cleavage reaction. Chemical reactions involve, for example, cleavage in acidic environments (e.g., endosomes), reductive cleavage (e.g., cytosolic cleavage) or oxidative cleavage (e.g., in liver microsomes). The cleavage reaction can also be initiated by a rearrangement reaction.

Enzymatic reactions can include reactions mediated by nucleases, peptidases, proteases, phosphatases, oxidases, reductases, etc. For example, a linker can be pH-sensitive, cathepsin- sensitive, or predominantly cleaved in endosomes and/or cytosol.

In some embodiments, the linker comprises a peptide. In certain embodiments, the linker comprises a peptide which includes a sequence that is cleavable by an endopeptidase. In addition to the cleavable peptide sequence, the linker may comprise additional amino acid residues and/or non-peptide chemical moieties, such as an alkyl chain. In certain

embodiments, the linker comprises Ala- Leu- Ala- Leu, which is a substrate for cathepsin B. See, for example, the maleimidocaproyl-Arg-Arg-Ala-Leu-Ala-Leu linkers described in

Schmid et al, Bioconjugate Chem 2007, 18, 702-716. In certain embodiments, a cathepsin B- cleavable linker is cleaved in tumor cells. In certain embodiments, the linker comprises Ala- Pro-Ile-Ser-Phe-Phe-Glu-Leu-Gly, which is a substrate for cathepsins D, L, and B (see, for example, Fischer et al, Chembiochem 2006, 7, 1428-1434). In certain embodiments, a cathepsin-cleavable linker is cleaved in HeLA cells. In some embodiments, the linker comprises Phe-Lys, which is a substrate for cathepsin B. For example, in certain

embodiments, the linker comprises Phe-Lys-p-aminobenzoic acid (PABA). See, e.g., the maleimidocaproyl-Phe-Lys-PABA linker described in Walker et al, Bioorg. Med. Chem. Lett. 2002, 12, 217-219. In certain embodiments, the linker comprises Gly-Phe-2- naphthylamide, which is a substrate for cathepsin C (see, for example, Berg et al. Biochem. J. 1994, 300, 229-235). In certain embodiments, a cathepsin C-cleavable linker is cleaved in hepatocytes. In some embodiments, the linker comprises a cathepsin S cleavage site. For example, in some embodiments, the linker comprises Gly-Arg-Trp-His-Thr-Val-Gly-Leu- Arg-Trp-Glu, Gly-Arg-Trp-Pro-Pro-Met-Gly-Leu-Pro-Trp-Glu, or Gly-Arg-Trp-His-Pro- Met-Gly-Ala-Pro-Trp-Glu, for example, as described in Lutzner et al, J. Biol. Chem. 2008, 283, 36185-36194. In certain embodiments, a cathepsin S-cleavable linker is cleaved in antigen presenting cells. In some embodiments, the linker comprises a papain cleavage site. Papain typically cleaves a peptide having the sequence -R/K-X-X (see Chapman et al, Annu. Rev. Physiol 1997, 59, 63-88). In certain embodiments, a papain-cleavable linker is cleaved in endosomes. In some embodiments, the linker comprises an endosomal acidic insulinase cleavage site. For example, in some embodiments, the linker comprises Tyr-Leu, Gly-Phe, or Phe-Phe (see, e.g., Authier et al, FEBS Lett. 1996, 389, 55-60). In certain embodiments, an endosomal acidic insulinase-cleavable linker is cleaved in hepatic cells.

In some embodiments, the linker is pH sensitive. In certain embodiments, the linker comprises a low pH-labile bond. As used herein, a low-pH labile bond is a bond that is selectively broken under acidic conditions (pH < 7). Such bonds may also be termed endosomally labile bonds, because cell endosomes and lysosomes have a pH less than 7. For example, in certain embodiments, the linker comprises an amine, an imine, an ester, a benzoic imine, an amino ester, a diortho ester, a polyphosphoester, a polyphosphazene, an acetal, a vinyl ether, a hydrazone, an azidomethyl-methylmaleic anhydride, a thiopropionate, a masked endosomolytic agent or a citraconyl group.

In certain embodiments, the linker comprises a low pH-labile bond selected from the following: ketals that are labile in acidic environments (e.g., pH less than 7, greater than about 4) to form a diol and a ketone; acetals that are labile in acidic environments (e.g., pH less than 7, greater than about 4) to form a diol and an aldehyde; imines or iminiums that are labile in acidic environments (e.g., pH less than 7, greater than about 4) to form an amine and an aldehyde or a ketone; silicon-oxygen-carbon linkages that are labile under acidic condition; silicon-nitrogne (silazane) linkages; silicon-carbon linkages (e.g., arylsilanes, vinylsilanes, and allylsilanes); maleamates (amide bonds synthesized from maleic anhydride derivatives and amines); ortho esters; hydrazones; activated carboxylic acid derivatives (e.g., esters, amides) designed to undergo acid catalyzed hydrolysis); or vinyl ethers. Further examples may be found in International Patent Appln. Pub. No. WO 2008/022309, entitled POLYCONJUGATES FOR IN VIVO DELIVERY OF POLYNUCLEOTIDES, the contents of which are incorporated herein by reference.

In some embodiments, the linker comprises a masked endosomolytic agent.

Endosomolytic polymers are polymers that, in response to a change in pH, are able to cause disruption or lysis of an endosome or provide for escape of a normally membrane- impermeable compound, such as a polynucleotide or protein, from a cellular internal membrane-enclosed vesicle, such as an endosome or lysosome. A subset of endosomolytic compounds is fusogenic compounds, including fusogenic peptides. Fusogenic peptides can facilitate endosomal release of agents such as oligomeric compounds to the cytoplasm. See, for example, US Patent Application Publication Nos. 20040198687, 20080281041,

20080152661, and 20090023890, which are incorporated herein by reference. The linker can also be designed to undergo an organ/ tissue-specific cleavage. For example, for certain targets, which are expressed in multiple tissues, only the knock-down in liver may be desirable, as knock-down in other organs may lead to undesired side effects. Thus, linkers susceptible to liver- specific enzymes, such as pyrrolase (TPO) and glucose-6- phosphatase (G-6-Pase), can be engineered, so as to limit the antisense effect to the liver mainly. Alternatively, linkers not susceptible to liver enzymes but susceptible to kidney- specific enzymes, such as gamma-glutamyltranspeptidase, can be engineered, so that the antisense effect is limited to the kidneys mainly. Analogously, intestine- specific peptidases cleaving Phe-Ala and Leu- Ala could be considered for orally administered multimeric oligonucleotides. Similarly, by placing an enzyme recognition site into the linker, which is recognized by an enzyme over-expressed in tumors, such as plasmin (e.g., PHEA-D-Val-Leu- Lys recognition site), tumor- specific knock-down should be feasible. By selecting the right enzyme recognition site in the linker, specific cleavage and knock-down should be achievable in many organs. In addition, the linker can also contain a targeting signal, such as N- acetyl galactosamine for liver targeting, or folate, vitamin A or RGD-peptide in the case of tumor or activated macrophage targeting. Accordingly, in some embodiments, the cleavable linker is organ- or tissue-specific, for example, liver- specific, kidney-specific, intestine- specific, etc.

The oligonucleotides can be linked through any part of the individual oligonucleotide, e.g., via the phosphate, the sugar (e.g., ribose, deoxyribose), or the nucleobase. In certain embodiments, when linking two oligonucleotides together, the linker can be attached e.g. to the 5 '-end of the first oligonucleotide and the 3 '-end of the second nucleotide, to the 5 '-end of the first oligonucleotide and the 5 'end of the second nucleotide, to the 3 '-end of the first oligonucleotide and the 3 '-end of the second nucleotide. In other embodiments, when linking two oligonucleotides together, the linker can attach internal residues of each oligonucleotides, e.g., via a modified nucleobase. One of ordinary skill in the art will understand that many such permutations are available for multimers. Further examples of appropriate linkers as well as methods for producing compounds having such linkers are disclosed in International Patent Application Number, PCT/US2012/055535, entitled MULTIMERIC

OLIGONUCLEOTIDE COMPOUNDS, publication number WO2013040429 Al, the contents of which relating to linkers and related chemistries are incorporated herein by referenced in its entirety. Methods for Selecting Candidate Oligonucleotides for Activating Expression of SMNl or SMN2

Methods are provided herein for selecting a candidate oligonucleotide for activating or enhancing expression of SMNl or SMN2. The target selection methods may generally involve steps for selecting single stranded oligonucleotides having any of the structural and functional characteristics disclosed herein. Typically, the methods involve one or more steps aimed at identifying oligonucleotides that target a PRC2-associated region that is functionally related to SMNl or SMN2, for example a PRC2-associated region of a IncRNA that regulates expression of SMNl or SMN2 by facilitating {e.g., in a -regulatory manner) the recruitment of PRC2 to the SMNl or SMN2 gene. Such oligonucleotides are expected to be candidates for activating expression of SMNl or SMN2 because of their ability to hybridize with the PRC2-associated region of a nucleic acid {e.g., a IncRNA). In some embodiments, this hybridization event is understood to disrupt interaction of PRC2 with the nucleic acid {e.g., a IncRNA) and as a result disrupt recruitment of PRC2 and its associated co-repressors {e.g., chromatin remodeling factors) to the SMNl or SMN2 gene locus.

Methods of selecting a candidate oligonucleotide may involve selecting a PRC2- associated region {e.g., a nucleotide sequence as set forth in any one of SEQ ID NOS: 9 to 29) that maps to a chromosomal position encompassing or in proximity to the SMNl or SMN2 gene {e.g., a chromosomal position having a sequence as set forth in any one of SEQ ID NOS: 1 to 8). The PRC2-associated region may map to the strand of the chromosome comprising the sense strand of the SMNl or SMN2 gene, in which case the candidate oligonucleotide is complementary to the sense strand of the SMNl or SMN2 gene {i.e., is antisense to the SMNl or SMN2 gene). Alternatively, the PRC2-associated region may map to the strand of the first chromosome comprising the antisense strand of the SMNl or SMN2 gene, in which case the oligonucleotide is complementary to the antisense strand (the template strand) of the SMNl or SMN2 gene {i.e., is sense to the SMNl or SMN2 gene).

Methods for selecting a set of candidate oligonucleotides that is enriched in oligonucleotides that activate expression of SMNl or SMN2 may involve selecting one or more PRC2-associated regions that map to a chromosomal position that encompasses or that is in proximity to the SMNl or SMN2 gene and selecting a set of oligonucleotides, in which each oligonucleotide in the set comprises a nucleotide sequence that is complementary with the one or more PRC2-associated regions. As used herein, the phrase, "a set of oligonucleotides that is enriched in oligonucleotides that activate expression of refers to a set of oligonucleotides that has a greater number of oligonucleotides that activate expression of a target gene (e.g. , SMN1 or SMN2) compared with a random selection of

oligonucleotides of the same physicochemical properties (e.g. , the same GC content, T m , length etc.) as the enriched set.

Where the design and/or synthesis of a single stranded oligonucleotide involves design and/or synthesis of a sequence that is complementary to a nucleic acid or PRC2- associated region described by such sequence information, the skilled person is readily able to determine the complementary sequence, e.g., through understanding of Watson Crick base pairing rules which form part of the common general knowledge in the field.

In some embodiments design and/or synthesis of a single stranded oligonucleotide involves manufacture of an oligonucleotide from starting materials by techniques known to those of skill in the art, where the synthesis may be based on a sequence of a PRC2- associated region, or portion thereof.

Methods of design and/or synthesis of a single stranded oligonucleotide may involve one or more of the steps of:

Identifying and/or selecting PRC2-associated region;

Designing a nucleic acid sequence having a desired degree of sequence identity or complementarity to a PRC2-associated region or a portion thereof;

Synthesizing a single stranded oligonucleotide to the designed sequence;

Purifying the synthesized single stranded oligonucleotide; and

Optionally mixing the synthesized single stranded oligonucleotide with at least one pharmaceutically acceptable diluent, carrier or excipient to form a pharmaceutical composition or medicament.

Single stranded oligonucleotides so designed and/or synthesized may be useful in method of modulating gene expression as described herein.

Preferably, single stranded oligonucleotides of the invention are synthesized chemically. Oligonucleotides used to practice this invention can be synthesized in vitro by well-known chemical synthesis techniques.

Oligonucleotides of the invention can be stabilized against nucleolytic degradation such as by the incorporation of a modification, e.g. , a nucleotide modification. For example, nucleic acid sequences of the invention include a phosphorothioate at least the first, second, or third internucleotide linkage at the 5' or 3' end of the nucleotide sequence. As another example, the nucleic acid sequence can include a 2'-modified nucleotide, e.g. , a 2'-deoxy, 2'- deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2'-0-MOE), 2'-0-aminopropyl (2'-0-AP), 2'-0-dimethylaminoethyl (2'-0-DMAOE), 2'-0-dimethylaminopropyl (2'-0-DMAP), 2'-0- dimethylaminoethyloxyethyl (2'-0-DMAEOE), or 2'-0-N-methylacetamido (2'-0-NMA). As another example, the nucleic acid sequence can include at least one 2'-0-methyl-modified nucleotide, and in some embodiments, all of the nucleotides include a 2'-0-methyl modification. In some embodiments, the nucleic acids are "locked," i.e., comprise nucleic acid analogues in which the ribose ring is "locked" by a methylene bridge connecting the 2'- O atom and the 4'-C atom.

It is understood that any of the modified chemistries or formats of single stranded oligonucleotides described herein can be combined with each other, and that one, two, three, four, five, or more different types of modifications can be included within the same molecule.

In some embodiments, the method may further comprise the steps of amplifying the synthesized single stranded oligonucleotide, and/or purifying the single stranded

oligonucleotide (or amplified single stranded oligonucleotide), and/or sequencing the single stranded oligonucleotide so obtained.

As such, the process of preparing a single stranded oligonucleotide may be a process that is for use in the manufacture of a pharmaceutical composition or medicament for use in the treatment of disease, optionally wherein the treatment involves modulating expression of a gene associated with a PRC2-associated region.

In the methods described above a PRC2-associated region may be, or have been, identified, or obtained, by a method that involves identifying RNA that binds to PRC2.

Such methods may involve the following steps: providing a sample containing nuclear ribonucleic acids, contacting the sample with an agent that binds specifically to PRC2 or a subunit thereof, allowing complexes to form between the agent and protein in the sample, partitioning the complexes, synthesizing nucleic acid that is complementary to nucleic acid present in the complexes.

Where the single stranded oligonucleotide is based on a PRC2-associated region, or a portion of such a sequence, it may be based on information about that sequence, e.g. , sequence information available in written or electronic form, which may include sequence information contained in publicly available scientific publications or sequence databases. In some embodiments, candidate oligonucleotides may be tested in animal models and/or cell-based models of ALS. Animal and cell-based models of ALS are known in the art. Exemplary animal models of ALS include SOD1 mutant transgenic mice (e.g.,

S OD 1 G93A mice) ^ S0D1 mutant tmns genie zebrafish, ALS2 knock-out mice, ALS2 knock- down zebrafish, transgenic human WT FUS mice, known down and transgenic mutant FUS zebrafish, TDP-43 knock out mice, transgenic mutant TDP-43 mice, transgenic mutant TDP- 43 zebrafish, transgenic mutant TDP-43 rats, Vps54 wr mice, Tbce^""" mice, Neurofilament light chain overexpressing transgenic mice, Neurofilament heavy chain overexpressing transgenic mice, and Peripherin overexpressing transgenic mice (see, e.g., Jakob Maximilian Moser, Paolo Bigini, and Thomas Schmitt-John. The wobbler mouse, an ALS animal model. Mol Genet Genomics. 2013; 288(5-6): 207-229; and Bruijn LI, Miller TM, Cleveland DW. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu Rev Neurosci. 2004;27:723-749). Exemplary cell-based models include cells (primary or cell- lines) derived from ALS patients, such as NSC-34/hSODl(G93A) cells and HeLa cells expressing FUS containing R495X ALS-causing mutation, or cells derived from an ALS animal model.

Nucleotide Analogues

In some embodiments, the oligonucleotide may comprise at least one ribonucleotide, at least one deoxyribonucleotide, and/or at least one bridged nucleotide. In some

embodiments, the oligonucleotide may comprise a bridged nucleotide, such as a locked nucleic acid (LNA) nucleotide, a constrained ethyl (cEt) nucleotide, or an ethylene bridged nucleic acid (ENA) nucleotide. Examples of such nucleotides are disclosed herein and known in the art. In some embodiments, the oligonucleotide comprises a nucleotide analog disclosed in one of the following United States Patent or Patent Application Publications: US 7,399,845, US 7,741,457, US 8,022,193, US 7,569,686, US 7,335,765, US 7,314,923, US 7,335,765, and US 7,816,333, US 20110009471, the entire contents of each of which are incorporated herein by reference for all purposes. The oligonucleotide may have one or more 2' O-methyl nucleotides. The oligonucleotide may consist entirely of 2' O-methyl nucleotides.

Often the single stranded oligonucleotide has one or more nucleotide analogues. For example, the single stranded oligonucleotide may have at least one nucleotide analogue that results in an increase in T m of the oligonucleotide in a range of 1°C, 2 °C, 3°C, 4 °C, or 5°C compared with an oligonucleotide that does not have the at least one nucleotide analogue. The single stranded oligonucleotide may have a plurality of nucleotide analogues that results in a total increase in T m of the oligonucleotide in a range of 2 °C, 3 °C, 4 °C, 5 °C, 6 °C, 7 °C, 8 °C, 9 °C, 10 °C, 15 °C, 20 °C, 25 °C, 30 °C, 35 °C, 40 °C, 45 °C or more compared with an oligonucleotide that does not have the nucleotide analogue.

The oligonucleotide may be of up to 50 nucleotides in length in which 2 to 10, 2 to 15 5 2 to 16, 2 to 17, 2 to 18, 2 to 19, 2 to 20, 2 to 25, 2 to 30, 2 to 40, 2 to 45, or more nucleotides of the oligonucleotide are nucleotide analogues. The oligonucleotide may be of 8 to 30 nucleotides in length in which 2 to 10, 2 to 15 5 2 to 16, 2 to 17, 2 to 18, 2 to 19, 2 to 20, 2 to 25, 2 to 30 nucleotides of the oligonucleotide are nucleotide analogues.

The oligonucleotide may be of 8 to 15 nucleotides in length in which 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, 2 to 10, 2 to 11, 2 to 12, 2 to 13, 2 to 14 nucleotides of the oligonucleotide are nucleotide analogues. Optionally, the oligonucleotides may have every nucleotide except 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides modified.

The oligonucleotide may consist entirely of bridged nucleotides (e.g. , LNA nucleotides, cEt nucleotides, ENA nucleotides). The oligonucleotide may comprise alternating deoxyribonucleotides and 2'-fluoro-deoxyribonucleotides. The oligonucleotide may comprise alternating deoxyribonucleotides and 2'-0-methyl nucleotides. The oligonucleotide may comprise alternating deoxyribonucleotides and ENA nucleotide analogues. The oligonucleotide may comprise alternating deoxyribonucleotides and LNA nucleotides. The oligonucleotide may comprise alternating LNA nucleotides and 2'-0- methyl nucleotides. The oligonucleotide may have a 5' nucleotide that is a bridged nucleotide (e.g. , a LNA nucleotide, cEt nucleotide, ENA nucleotide). The oligonucleotide may have a 5' nucleotide that is a deoxyribonucleotide.

The oligonucleotide may comprise deoxyribonucleotides flanked by at least one bridged nucleotide (e.g. , a LNA nucleotide, cEt nucleotide, ENA nucleotide) on each of the 5' and 3' ends of the deoxyribonucleotides. The oligonucleotide may comprise

deoxyribonucleotides flanked by 1, 2, 3, 4, 5, 6, 7, 8 or more bridged nucleotides (e.g. , LNA nucleotides, cEt nucleotides, ENA nucleotides) on each of the 5' and 3' ends of the deoxyribonucleotides. The 3' position of the oligonucleotide may have a 3' hydroxyl group. The 3' position of the oligonucleotide may have a 3' thiophosphate. The oligonucleotide may be conjugated with a label. For example, the oligonucleotide may be conjugated with a biotin moiety, cholesterol, Vitamin A, folate, sigma receptor ligands, aptamers, peptides, such as CPP, hydrophobic molecules, such as lipids, ASGPR or dynamic polyconjugates and variants thereof at its 5' or 3' end.

Preferably the single stranded oligonucleotide comprises one or more modifications comprising: a modified sugar moiety, and/or a modified internucleoside linkage, and/or a modified nucleotide and/or combinations thereof. It is not necessary for all positions in a given oligonucleotide to be uniformly modified, and in fact more than one of the

modifications described herein may be incorporated in a single oligonucleotide or even at within a single nucleoside within an oligonucleotide.

In some embodiments, the single stranded oligonucleotides are chimeric

oligonucleotides that contain two or more chemically distinct regions, each made up of at least one nucleotide. These oligonucleotides typically contain at least one region of modified nucleotides that confers one or more beneficial properties (such as, for example, increased nuclease resistance, increased uptake into cells, increased binding affinity for the target) and a region that is a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. Chimeric single stranded oligonucleotides of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides,

oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures comprise, but are not limited to, US patent nos. 5,013,830; 5,149,797; 5, 220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133;

5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, each of which is herein incorporated by reference.

In some embodiments, the single stranded oligonucleotide comprises at least one nucleotide modified at the 2' position of the sugar, most preferably a 2'-0-alkyl, 2'-0-alkyl-0- alkyl or 2'-fluoro-modified nucleotide. In other preferred embodiments, RNA modifications include 2'-fluoro, 2'-amino and 2' O-methyl modifications on the ribose of pyrimidines, abasic residues or an inverted base at the 3' end of the RNA. Such modifications are routinely incorporated into oligonucleotides and these oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than 2'-deoxyoligonucleotides against a given target. A number of nucleotide and nucleoside modifications have been shown to make the oligonucleotide into which they are incorporated more resistant to nuclease digestion than the native oligodeoxynucleotide; these modified oligos survive intact for a longer time than unmodified oligonucleotides. Specific examples of modified oligonucleotides include those comprising modified backbones, for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages. Most preferred are oligonucleotides with

phosphorothioate backbones and those with heteroatom backbones, particularly CH 2 -NH-O- CH 2 , CH,~N(CH 3 )~0~CH 2 (known as a methylene(methylimino) or MMI backbone, CH 2 - O-N (CH 3 )-CH 2 , CH 2 -N (CH 3 )-N (CH 3 )-CH 2 and O-N (CH 3 )- CH 2 -CH 2 backbones, wherein the native phosphodiester backbone is represented as O- P— O- CH,); amide backbones (see De Mesmaeker et al. Ace. Chem. Res. 1995, 28:366-374); morpholino backbone structures (see Summerton and Weller, U.S. Pat. No. 5,034,506); peptide nucleic acid (PNA) backbone (wherein the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleotides being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone, see Nielsen et al., Science 1991, 254, 1497). Phosphorus-containing linkages include, but are not limited to, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates comprising 3'alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates comprising 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3 -5' to 5'-3' or 2 -5' to 5'-2'; see US patent nos. 3,687,808; 4,469,863;

4,476,301; 5,023,243; 5, 177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455, 233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563, 253; 5,571,799; 5,587,361; and 5,625,050.

Morpholino-based oligomeric compounds are described in Dwaine A. Braasch and David R. Corey, Biochemistry, 2002, 41(14), 4503-4510); Genesis, volume 30, issue 3, 2001; Heasman, J., Dev. Biol., 2002, 243, 209-214; Nasevicius et al., Nat. Genet., 2000, 26, 216- 220; Lacerra et al., Proc. Natl. Acad. Sci., 2000, 97, 9591-9596; and U.S. Pat. No. 5,034,506, issued Jul. 23, 1991. In some embodiments, the morpholino-based oligomeric compound is a phosphorodiamidate morpholino oligomer (PMO) (e.g. , as described in Iverson, Curr. Opin. Mol. Ther., 3:235-238, 2001 ; and Wang et al., J. Gene Med., 12:354-364, 2010; the disclosures of which are incorporated herein by reference in their entireties).

Cyclohexenyl nucleic acid oligonucleotide mimetics are described in Wang et al., J. Am. Chem. Soc, 2000, 122, 8595-8602.

Modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These comprise those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts; see US patent nos. 5,034,506; 5, 166,315; 5,185,444; 5,214,134; 5,216, 141 ; 5,235,033; 5,264, 562; 5, 264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596, 086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623, 070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439, each of which is herein incorporated by reference.

Modified oligonucleotides are also known that include oligonucleotides that are based on or constructed from arabinonucleotide or modified arabinonucleotide residues.

Arabinonucleosides are stereoisomers of ribonucleosides, differing only in the configuration at the 2'-position of the sugar ring. In some embodiments, a 2'-arabino modification is 2'-F arabino. In some embodiments, the modified oligonucleotide is 2' -fluoro-D-arabinonucleic acid (FANA) (as described in, for example, Lon et al., Biochem., 41 :3457-3467, 2002 and Min et al., Bioorg. Med. Chem. Lett., 12:2651-2654, 2002; the disclosures of which are incorporated herein by reference in their entireties). Similar modifications can also be made at other positions on the sugar, particularly the 3' position of the sugar on a 3' terminal nucleoside or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide.

PCT Publication No. WO 99/67378 discloses arabinonucleic acids (ANA) oligomers and their analogues for improved sequence specific inhibition of gene expression via association to complementary messenger RNA. Other preferred modifications include ethylene-bridged nucleic acids (ENAs) (e.g. , International Patent Publication No. WO 2005/042777, Morita et al., Nucleic Acid Res., Suppl 1 :241-242, 2001 ; Surono et al., Hum. Gene Ther., 15:749-757, 2004; Koizumi, Curr. Opin. Mol. Ther., 8: 144- 149, 2006 and Horie et al., Nucleic Acids Symp. Ser (Oxf), 49: 171- 172, 2005; the disclosures of which are incorporated herein by reference in their entireties). Preferred ENAs include, but are not limited to, 2'-0,4'-C-ethylene -bridged nucleic acids.

Examples of LNAs are described in WO/2008/043753 and include compounds of the following general formula.

where X and Y are independently selected among the groups -0-,

-S-, -N(H)-, N(Pv)-, -CH 2 - or -CH- (if part of a double bond),

-CH 2 -0-, -CH 2 -S-, -CH 2 -N(H)-, -CH 2 -N(R)-, -CH 2 -CH 2 - or -CH 2 -CH- (if part of a double bond),

-CH=CH-, where R is selected from hydrogen and Ci-4-alkyl; Z and Z* are independently selected among an internucleoside linkage, a terminal group or a protecting group; B constitutes a natural or non-natural nucleotide base moiety; and the asymmetric groups may be found in either orientation. Preferably, the LNA used in the oligonucleotides described herein comprises at least one LNA unit according any of the formulas wherein Y is -0-, -S-, -NH-, or N(R ); Z and Z* are independently selected among an intemucleoside linkage, a terminal group or a protecting group; B constitutes a natural or non-natural nucleotide base moiety, and RH is selected from hydrogen and Ci-4-alkyl.

In some embodiments, the Locked Nucleic Acid (LNA) used in the oligonucleotides described herein comprises at least one Locked Nucleic Acid (LNA) unit according any of the formulas shown in Scheme 2 of PCT/DK2006/000512.

In some embodiments, the LNA used in the oligomer of the invention comprises intemucleoside linkages selected from -0-P(O) 2 -O-, -0-P(0,S)-0-, -0-P(S) 2 -O-, -S-P(0) 2 -0-, -S-P(0,S)-0-, -S-P(S) 2 -0-, -0-P(O) 2 -S-, -0-P(0,S)-S-, -S-P(0) 2 -S-, -0-PO(R H )-0-, o- PO(OCH 3 )-0-, -0-PO(NR H )-0-, -0-PO(OCH 2 CH 2 S-R)-O-, -0-PO(BH 3 )-0-, -0-PO(NHR H )- 0-, -0-P(0) 2 -NR H -, -NR H -P(0) 2 -0-, -NR H -CO-0-, where R H is selected from hydrogen and Ci_4-alkyl.

Specifically preferred LNA units are shown in scheme 2:

-O-thio-tNA

p-D-E A

»D*amSrto » Li¾A

Scheme 2

The term "thio-LNA" comprises a locked nucleotide in which at least one of X or Y in the general formula above is selected from S or -CH 2 -S-. Thio-LNA can be in both beta-D and alpha-L-configuration.

The term "amino-LNA" comprises a locked nucleotide in which at least one of X or Y in the general formula above is selected from -N(H)-, N(R)-, CH 2 -N(H)-, and -CH 2 -N(R)- where R is selected from hydrogen and Ci-4-alkyl. Amino-LNA can be in both beta-D and alpha-L-configuration.

The term "oxy-LNA" comprises a locked nucleotide in which at least one of X or Y in the general formula above represents -O- or -CH 2 -0-. Oxy-LNA can be in both beta-D and alpha-L-configuration. The term "ena-LNA" comprises a locked nucleotide in which Y in the general formula above is -CH 2 -0- (where the oxygen atom of -CH 2 -0- is attached to the 2'-position relative to the base B).

LNAs are described in additional detail herein.

One or more substituted sugar moieties can also be included, e.g. , one of the following at the 2' position: OH, SH, SCH 3 , F, OCN, OCH 3 OCH 3 , OCH 3 0(CH 2 )n CH 3 , 0(CH 2 )n NH 2 or 0(CH 2 )n CH 3 where n is from 1 to about 10; CI to CIO lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; CI; Br; CN; CF ; OCF ; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; ON0 2 ; N0 2 ; N 3 ; NH2; heterocycloalkyl; heterocyclo alkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A preferred modification includes 2'-methoxyethoxy [2'-0-CH 2 CH 2 OCH 3 , also known as 2'-0-(2-methoxyethyl)] (Martin et al, Helv. Chim. Acta, 1995, 78, 486). Other preferred modifications include 2'- methoxy (2'-0-CH 3 ), 2'-propoxy (2'-OCH 2 CH 2 CH 3 ) and 2'-fluoro (2'-F). Similar

modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group.

Single stranded oligonucleotides can also include, additionally or alternatively, nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include adenine (A), guanine (G), thymine (T), cytosine (C) and uracil (U). Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e.g. , hypoxanthine, 6-methyladenine, 5-Me pyrimidines, particularly 5-methylcytosine (also referred to as 5-methyl-2'

deoxycytosine and often referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC and gentobiosyl HMC, isocytosine, pseudoisocytosine, as well as synthetic nucleobases, e.g. , 2-aminoadenine, 2- (methylamino)adenine, 2-(imidazolylalkyl)adenine, 2- (aminoalklyamino)adenine or other hetero substituted alkyladenines, 2-thiouracil, 2- thiothymine, 5-bromouracil, 5-hydroxymethyluracil, 5-propynyluracil, 8-azaguanine, 7- deazaguanine, N6 (6-aminohexyl)adenine, 6-aminopurine, 2-aminopurine, 2-chloro-6- aminopurine and 2,6-diaminopurine or other diaminopurines. See, e.g. , Kornberg, "DNA Replication," W. H. Freeman & Co., San Francisco, 1980, pp75-77; and Gebeyehu, G., et al. Nucl. Acids Res., 15:4513 (1987)). A "universal" base known in the art, e.g. , inosine, can also be included. 5-Me-C substitutions have been shown to increase nucleic acid duplex stability by 0.6- 1.2°C. (Sanghvi, in Crooke, and Lebleu, eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and may be used as base substitutions.

It is not necessary for all positions in a given oligonucleotide to be uniformly modified, and in fact more than one of the modifications described herein may be

incorporated in a single oligonucleotide or even at within a single nucleoside within an oligonucleotide.

In some embodiments, both a sugar and an internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar- backbone of an oligonucleotide is replaced with an amide containing backbone, for example, an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, US patent nos. 5,539,082; 5,714,331 ; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al, Science, 1991, 254, 1497-1500.

Single stranded oligonucleotides can also include one or more nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases comprise the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified

nucleobases comprise other synthetic and natural nucleobases such as 5-methylcytosine (5- me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudo-uracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8- thioalkyl, 8-hydroxyl and other 8- substituted adenines and guanines, 5-halo particularly 5- bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylquanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3- deazaguanine and 3-deazaadenine.

Further, nucleobases comprise those disclosed in United States Patent No. 3,687,808, those disclosed in "The Concise Encyclopedia of Polymer Science And Engineering", pages 858-859, Kroschwitz, ed. John Wiley & Sons, 1990;, those disclosed by Englisch et al., Angewandle Chemie, International Edition, 1991, 30, page 613, and those disclosed by Sanghvi, Chapter 15, Antisense Research and Applications," pages 289- 302, Crooke, and Lebleu, eds., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, comprising 2-aminopropyladenine, 5-propynyluracil and 5- propynylcytosine. 5- methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6- 1.2<0>C (Sanghvi, et al., eds, "Antisense Research and Applications," CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2'-0-methoxyethyl sugar modifications. Modified nucleobases are described in US patent nos. 3,687,808, as well as 4,845,205; 5,130,302; 5,134,066; 5, 175, 273; 5, 367,066; 5,432,272; 5,457, 187; 5,459,255; 5,484,908; 5,502,177; 5,525,711 ; 5,552,540; 5,587,469; 5,596,091 ; 5,614,617; 5,750,692, and 5,681,941, each of which is herein incorporated by reference.

In some embodiments, the single stranded oligonucleotides are chemically linked to one or more moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide. For example, one or more single stranded oligonucleotides, of the same or different types, can be conjugated to each other; or single stranded

oligonucleotides can be conjugated to targeting moieties with enhanced specificity for a cell type or tissue type. Such moieties include, but are not limited to, lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g. , hexyl-S- tritylthiol (Manoharan et al, Ann. N. Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g. , dodecandiol or undecyl residues (Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimie, 1993, 75, 49- 54), a phospholipid, e.g. , di-hexadecyl-rac-glycerol or triethylammonium 1,2- di-O-hexadecyl- rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Mancharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-t oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937). See also US patent nos. 4,828,979; 4,948,882; 5,218, 105; 5,525,465; 5,541,313; 5,545,730; 5,552, 538; 5,578,717, 5,580,731 ; 5,580,731 ; 5,591,584; 5,109, 124; 5, 118,802; 5,138,045; 5,414,077; 5,486, 603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762, 779; 4,789,737; 4,824,941 ; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082, 830; 5,112,963; 5,214,136; 5,082,830; 5, 112,963; 5,214, 136; 5, 245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391, 723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5, 565,552; 5,567,810; 5,574, 142; 5,585,481 ; 5,587,371 ; 5,595,726; 5,597,696; 5,599,923; 5,599, 928 and 5,688,941, each of which is herein incorporated by reference.

These moieties or conjugates can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups. Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence- specific hybridization with the target nucleic acid. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups that improve uptake, distribution, metabolism or excretion of the compounds of the present invention. Representative conjugate groups are disclosed in International Patent Application No. PCT/US92/09196, filed Oct. 23, 1992, and U.S. Pat. No. 6,287,860, which are incorporated herein by reference. Conjugate moieties include, but are not limited to, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g. , hexyl-5-tritylthiol, a thiocholesterol, an aliphatic chain, e.g. , dodecandiol or undecyl residues, a phospholipid, e.g. , di-hexadecyl-rac- glycerol or triethylammonium 1,2- di-O-hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxy cholesterol moiety. See, e.g. , U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218, 105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731 ; 5,580,731 ; 5,591,584; 5,109, 124; 5,118,802; 5, 138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941 ; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5, 112,963; 5,214, 136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574, 142; 5,585,481 ; 5,587,371 ; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941.

In some embodiments, single stranded oligonucleotide modification include modification of the 5' or 3' end of the oligonucleotide. In some embodiments, the 3' end of the oligonucleotide comprises a hydroxyl group or a thiophosphate. It should be appreciated that additional molecules (e.g. a biotin moiety or a fluorophor) can be conjugated to the 5' or 3' end of the single stranded oligonucleotide. In some embodiments, the single stranded oligonucleotide comprises a biotin moiety conjugated to the 5' nucleotide.

In some embodiments, the single stranded oligonucleotide comprises locked nucleic acids (LNA), ENA modified nucleotides, 2'-0-methyl nucleotides, or 2'-fluoro- deoxyribonucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and 2'-fluoro-deoxyribonucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and 2'-0- methyl nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and ENA modified nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and locked nucleic acid nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating locked nucleic acid nucleotides and 2'-0-methyl nucleotides.

In some embodiments, the 5' nucleotide of the oligonucleotide is a

deoxyribonucleotide. In some embodiments, the 5' nucleotide of the oligonucleotide is a locked nucleic acid nucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one locked nucleic acid nucleotide on each of the 5' and 3' ends of the deoxyribonucleotides. In some embodiments, the nucleotide at the 3' position of the oligonucleotide has a 3' hydroxyl group or a 3' thiophosphate.

In some embodiments, the single stranded oligonucleotide comprises

phosphorothioate internucleotide linkages. In some embodiments, the single stranded oligonucleotide comprises phosphorothioate internucleotide linkages between at least two nucleotides. In some embodiments, the single stranded oligonucleotide comprises phosphorothioate internucleotide linkages between all nucleotides.

It should be appreciated that the single stranded oligonucleotide can have any combination of modifications as described herein.

The oligonucleotide may comprise a nucleotide sequence having one or more of the following modification patterns.

(a) (X)Xxxxxx, (X)xXxxxx, (X)xxXxxx, (X)xxxXxx, (X)xxxxXx and (X)xxxxxX,

(b) (X)XXxxxx, (X)XxXxxx, (X)XxxXxx, (X)XxxxXx, (X)XxxxxX, (X)xXXxxx, (X)xXxXxx, (X)xXxxXx, (X)xXxxxX, (X)xxXXxx, (X)xxXxXx, (X)xxXxxX, (X)xxxXXx,

(X)xxxXxX and (X)xxxxXX,

(c) (X)XXXxxx, (X)xXXXxx, (X)xxXXXx, (X)xxxXXX, (X)XXxXxx, (X)XXxxXx, (X)XXxxxX, (X)xXXxXx, (X)xXXxxX, (X)xxXXxX, (X)XxXXxx, (X)XxxXXx

(X)XxxxXX, (X)xXxXXx, (X)xXxxXX, (X)xxXxXX, (X)xXxXxX and (X)XxXxXx,

(d) (X)xxXXX, (X)xXxXXX, (X)xXXxXX, (X)xXXXxX, (X)xXXXXx,

(X)XxxXXXX, (X)XxXxXX, (X)XxXXxX, (X)XxXXx, (X)XXxxXX, (X)XXxXxX, (X)XXxXXx, (X)XXXxxX, (X)XXXxXx, and (X)XXXXxx,

(e) (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXXxX and (X)XXXXXx, and

(f) XXXXXX, XxXXXXX, XXxXXXX, XXXxXXX, XXXXxXX, XXXXXxX and

XXXXXXx, in which "X" denotes a nucleotide analogue, (X) denotes an optional nucleotide analogue, and "x" denotes a DNA or RNA nucleotide unit. Each of the above listed patterns may appear one or more times within an oligonucleotide, alone or in combination with any of the other disclosed modification patterns. Methods for Modulating Gene Expression

In some embodiments, methods are provided for increasing expression of SMN protein in a cell. The methods, in some embodiments, involve delivering to the cell a first single stranded oligonucleotide complementary with a PRC2-associated region of SMNl or SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMNl or SMN2, in amounts sufficient to increase expression of a mature mRNA of SMNl or SMN2 that comprises (or includes) exon 7 in the cell. The first and second single stranded oligonucleotides may be delivered together or separately. The first and second single stranded oligonucleotides may be linked together, or unlinked, i.e., separate.

In some embodiments, methods are provided for treating ALS in a subject. The methods, in some embodiments, involve administering to a subject a first single stranded oligonucleotide complementary with a PRC2-associated region of SMNl or SMN2 and a second single stranded oligonucleotide complementary with a splice control sequence of a precursor mRNA of SMNl or SMN2, in amounts sufficient to increase expression of full length SMN protein in the subject to levels sufficient to improve one or more conditions associated with ALS. The first and second single stranded oligonucleotides may be administered together or separately. The first and second single stranded oligonucleotides may be linked together, or unlinked, i.e., separate. The first single stranded oligonucleotide may be administered within 1 hour, 2 hours, 3 hours, 4 hours, 8 hours, 12 hours, 24 hours, 48 hours or more of administration of the second single stranded oligonucleotide. The first single stranded oligonucleotide may be administered before or after the second single stranded oligonucleotide. The oligonucleotides may be administered once or on multiple occasions depending on the needs of the subject and/or judgment of the treating physician. In some cases, the oligonucleotides may be administered in cycles. The administration cycles may vary; for example, the administration cycle may be 2 nd oligo - 1 st oligo - 2 nd oligo - 1 st oligo and so on; or 1 st oligo-2 nd oligo- 1 st oligo-2 nd oligo, and so on; or 1 st oligo - 2 nd oligo - 2 nd oligo -1 st oligo- 1 st oligo - 2 nd oligo - 2 nd oligo -1 st oligo, and so on. The skilled artisan will be capable of selecting administration cycles and intervals between each administration that are appropriate for treating a particular subject.

In one aspect, the invention relates to methods for modulating gene expression in a cell (e.g., a cell for which SMNl or SMN2 levels are reduced) for research purposes (e.g., to study the function of the gene in the cell). In another aspect, the invention relates to methods for modulating gene expression in a cell (e.g. , a cell for which SMNl or SMN2 levels are reduced) for gene or epigenetic therapy. The cells can be in vitro, ex vivo, or in vivo (e.g. , in a subject who has a disease resulting from reduced expression or activity of SMNl or SMN2. In some embodiments, methods for modulating gene expression in a cell comprise delivering a single stranded oligonucleotide as described herein. In some embodiments, delivery of the single stranded oligonucleotide to the cell results in a level of expression of gene that is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200% or more greater than a level of expression of gene in a control cell to which the single stranded

oligonucleotide has not been delivered. In certain embodiments, delivery of the single stranded oligonucleotide to the cell results in a level of expression of gene that is at least 50% greater than a level of expression of gene in a control cell to which the single stranded oligonucleotide has not been delivered.

In another aspect of the invention, methods comprise administering to a subject (e.g. a human) a composition comprising a single stranded oligonucleotide as described herein to increase protein levels in the subject. In some embodiments, the increase in protein levels is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, or more, higher than the amount of a protein in the subject before administering.

As another example, to increase expression of SMNl or SMN2 in a cell, the methods include introducing into the cell a single stranded oligonucleotide that is sufficiently complementary to a PRC2-associated region (e.g. , of a long non-coding RNA) that maps to a genomic position encompassing or in proximity to the SMNl or SMN2 gene.

In another aspect of the invention provides methods of treating a condition (e.g. , ALS) associated with decreased levels of expression or activity of SMNl or SMN2 in a subject, the method comprising administering a single stranded oligonucleotide as described herein.

A subject can include a non-human mammal, e.g. mouse, rat, guinea pig, rabbit, cat, dog, goat, cow, or horse. In preferred embodiments, a subject is a human. Single stranded oligonucleotides have been employed as therapeutic moieties in the treatment of disease states in animals, including humans. Single stranded oligonucleotides can be useful therapeutic modalities that can be configured to be useful in treatment regimes for the treatment of cells, tissues and animals, especially humans. For therapeutics, an animal, preferably a human, suspected of having ALSis treated by administering single stranded oligonucleotide in accordance with this invention. For example, in one non-limiting embodiment, the methods comprise the step of administering to the animal in need of treatment, a therapeutically effective amount of a single stranded oligonucleotide as described herein.

Formulation, Delivery, And Dosing

The oligonucleotides described herein can be formulated for administration to a subject for treating a condition (e.g., ALS) associated with decreased levels or activity of SMN protein. It should be understood that the formulations, compositions and methods can be practiced with any of the oligonucleotides disclosed herein. In some embodiments, formulations are provided that comprise a first single stranded oligonucleotide

complementary with a PRC2-associated region of a gene and a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, formulations are provided that comprise a first single stranded oligonucleotide complementary with a PRC2-associated region of a gene that is linked via a linker with a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. Thus, it should be appreciated that in some embodiments, a first single stranded oligonucleotide complementary with a PRC2-associated region of a gene is linked with a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene, and in other embodiments, the single stranded oligonucleotides are not linked. Single stranded oligonucleotides that are not linked may be administered to a subject or delivered to a cell simultaneously (e.g., within the same composition) or separately.

The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient (e.g., an oligonucleotide or compound of the invention) which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration, e.g., intradermal or inhalation. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect, e.g. tumor regression. Pharmaceutical formulations of this invention can be prepared according to any method known to the art for the manufacture of pharmaceuticals. Such formulations can contain sweetening agents, flavoring agents, coloring agents and preserving agents. A formulation can be admixtured with nontoxic pharmaceutically acceptable excipients which are suitable for manufacture. Formulations may comprise one or more diluents, emulsifiers, preservatives, buffers, excipients, etc. and may be provided in such forms as liquids, powders, emulsions, lyophilized powders, sprays, creams, lotions, controlled release formulations, tablets, pills, gels, on patches, in implants, etc.

A formulated single stranded oligonucleotide composition can assume a variety of states. In some examples, the composition is at least partially crystalline, uniformly crystalline, and/or anhydrous (e.g. , less than 80, 50, 30, 20, or 10% water). In another example, the single stranded oligonucleotide is in an aqueous phase, e.g. , in a solution that includes water. The aqueous phase or the crystalline compositions can, e.g. , be incorporated into a delivery vehicle, e.g. , a liposome (particularly for the aqueous phase) or a particle (e.g. , a microparticle as can be appropriate for a crystalline composition). Generally, the single stranded oligonucleotide composition is formulated in a manner that is compatible with the intended method of administration.

In some embodiments, the composition is prepared by at least one of the following methods: spray drying, lyophilization, vacuum drying, evaporation, fluid bed drying, or a combination of these techniques; or sonication with a lipid, freeze-drying, condensation and other self-assembly.

A single stranded oligonucleotide preparation can be formulated or administered (together or separately) in combination with another agent, e.g. , another therapeutic agent or an agent that stabilizes a single stranded oligonucleotide, e.g. , a protein that complexes with single stranded oligonucleotide. Still other agents include chelators, e.g. , EDTA (e.g. , to remove divalent cations such as Mg 2+ ), salts, RNAse inhibitors (e.g. , a broad specificity RNAse inhibitor such as RNAsin) and so forth. In some embodiments, the other agent used in combination with the single stranded oligonucleotide is an agent that also regulates SMN expression. In some embodiments, the other agent is a growth hormone, a histone

deacetylase inhibitor, a hydroxycarbamide (hydroxyurea), a natural polyphenol compound (e.g., resveratrol, curcumin), prolactin, or salbutamol. Examples of histone deacetylase inhibitors that may be used include aliphatic compounds (e.g., butyrates (e.g., sodium butyrate and sodium phenylbutyrate) and valproic acid), benzamides (e.g., M344), and hydroxamic acids (e.g., CBHA, SBHA, Entinostat (MS-275)) Panobinostat (LBH-589), Trichostatin A, Vorinostat (SAHA)),

In one embodiment, the single stranded oligonucleotide preparation includes another single stranded oligonucleotide, e.g., a second single stranded oligonucleotide that modulates expression and/or mRNA processing of a second gene or a second single stranded

oligonucleotide that modulates expression of the first gene. Still other preparation can include at least 3, 5, ten, twenty, fifty, or a hundred or more different single stranded oligonucleotide species. Such single stranded oligonucleotides can mediated gene expression with respect to a similar number of different genes. In one embodiment, the single stranded oligonucleotide preparation includes at least a second therapeutic agent (e.g., an agent other than an oligonucleotide).

Route of Delivery

A composition that includes a single stranded oligonucleotide can be delivered to a subject by a variety of routes. Exemplary routes include: intravenous, intradermal, topical, rectal, parenteral, anal, intravaginal, intranasal, pulmonary, ocular. The term "therapeutically effective amount" is the amount of oligonucleotide present in the composition that is needed to provide the desired level of SMNl or SMN2 expression in the subject to be treated to give the anticipated physiological response. The term "physiologically effective amount" is that amount delivered to a subject to give the desired palliative or curative effect. The term "pharmaceutically acceptable carrier" means that the carrier can be administered to a subject with no significant adverse toxicological effects to the subject.

The single stranded oligonucleotide molecules of the invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically include one or more species of single stranded oligonucleotide and a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.

The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic, vaginal, rectal, intranasal, transdermal), oral or parenteral. Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, or intrathecal or

intraventricular administration.

The route and site of administration may be chosen to enhance targeting. For example, to target muscle cells, intramuscular injection into the muscles of interest would be a logical choice. Lung cells might be targeted by administering the single stranded oligonucleotide in aerosol form. The vascular endothelial cells could be targeted by coating a balloon catheter with the single stranded oligonucleotide and mechanically introducing the oligonucleotide.

Topical administration refers to the delivery to a subject by contacting the formulation directly to a surface of the subject. The most common form of topical delivery is to the skin, but a composition disclosed herein can also be directly applied to other surfaces of the body, e.g. , to the eye, a mucous membrane, to surfaces of a body cavity or to an internal surface. As mentioned above, the most common topical delivery is to the skin. The term encompasses several routes of administration including, but not limited to, topical and transdermal. These modes of administration typically include penetration of the skin's permeability barrier and efficient delivery to the target tissue or stratum. Topical administration can be used as a means to penetrate the epidermis and dermis and ultimately achieve systemic delivery of the composition. Topical administration can also be used as a means to selectively deliver oligonucleotides to the epidermis or dermis of a subject, or to specific strata thereof, or to an underlying tissue.

Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful.

Transdermal delivery is a valuable route for the administration of lipid soluble therapeutics. The dermis is more permeable than the epidermis and therefore absorption is much more rapid through abraded, burned or denuded skin. Inflammation and other physiologic conditions that increase blood flow to the skin also enhance transdermal adsorption. Absorption via this route may be enhanced by the use of an oily vehicle

(inunction) or through the use of one or more penetration enhancers. Other effective ways to deliver a composition disclosed herein via the transdermal route include hydration of the skin and the use of controlled release topical patches. The transdermal route provides a potentially effective means to deliver a composition disclosed herein for systemic and/or local therapy. In addition, iontophoresis (transfer of ionic solutes through biological membranes under the influence of an electric field), phonophoresis or sonophoresis (use of ultrasound to enhance the absorption of various therapeutic agents across biological membranes, notably the skin and the cornea), and optimization of vehicle characteristics relative to dose position and retention at the site of administration may be useful methods for enhancing the transport of topically applied compositions across skin and mucosal sites.

Both the oral and nasal membranes offer advantages over other routes of

administration. For example, oligonucleotides administered through these membranes may have a rapid onset of action, provide therapeutic plasma levels, avoid first pass effect of hepatic metabolism, and avoid exposure of the oligonucleotides to the hostile gastrointestinal (GI) environment. Additional advantages include easy access to the membrane sites so that the oligonucleotide can be applied, localized and removed easily.

In oral delivery, compositions can be targeted to a surface of the oral cavity, e.g. , to sublingual mucosa which includes the membrane of ventral surface of the tongue and the floor of the mouth or the buccal mucosa which constitutes the lining of the cheek. The sublingual mucosa is relatively permeable thus giving rapid absorption and acceptable bioavailability of many agents. Further, the sublingual mucosa is convenient, acceptable and easily accessible.

A pharmaceutical composition of single stranded oligonucleotide may also be administered to the buccal cavity of a human being by spraying into the cavity, without inhalation, from a metered dose spray dispenser, a mixed micellar pharmaceutical

formulation as described above and a propellant. In one embodiment, the dispenser is first shaken prior to spraying the pharmaceutical formulation and propellant into the buccal cavity.

Compositions for oral administration include powders or granules, suspensions or solutions in water, syrups, slurries, emulsions, elixirs or non-aqueous media, tablets, capsules, lozenges, or troches. In the case of tablets, carriers that can be used include lactose, sodium citrate and salts of phosphoric acid. Various disintegrants such as starch, and lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc, are commonly used in tablets. For oral administration in capsule form, useful diluents are lactose and high molecular weight polyethylene glycols. When aqueous suspensions are required for oral use, the nucleic acid compositions can be combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents can be added.

Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, intrathecal or intraventricular administration. In some embodiments, parental administration involves administration directly to the site of disease (e.g. injection into a tumor).

Formulations for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir. For intravenous use, the total concentration of solutes should be controlled to render the preparation isotonic.

Any of the single stranded oligonucleotides described herein can be administered to ocular tissue. For example, the compositions can be applied to the surface of the eye or nearby tissue, e.g. , the inside of the eyelid. For ocular administration, ointments or droppable liquids may be delivered by ocular delivery systems known to the art such as applicators or eye droppers. Such compositions can include mucomimetics such as hyaluronic acid, chondroitin sulfate, hydroxypropyl methylcellulose or poly(vinyl alcohol), preservatives such as sorbic acid, EDTA or benzylchronium chloride, and the usual quantities of diluents and/or carriers. The single stranded oligonucleotide can also be administered to the interior of the eye, and can be introduced by a needle or other delivery device which can introduce it to a selected area or structure.

Pulmonary delivery compositions can be delivered by inhalation by the patient of a dispersion so that the composition, preferably single stranded oligonucleotides, within the dispersion can reach the lung where it can be readily absorbed through the alveolar region directly into blood circulation. Pulmonary delivery can be effective both for systemic delivery and for localized delivery to treat diseases of the lungs. Pulmonary delivery can be achieved by different approaches, including the use of nebulized, aerosolized, micellular and dry powder-based formulations. Delivery can be achieved with liquid nebulizers, aerosol-based inhalers, and dry powder dispersion devices. Metered-dose devices are preferred. One of the benefits of using an atomizer or inhaler is that the potential for contamination is minimized because the devices are self-contained. Dry powder dispersion devices, for example, deliver agents that may be readily formulated as dry powders. A single stranded oligonucleotide composition may be stably stored as lyophilized or spray-dried powders by itself or in combination with suitable powder carriers. The delivery of a composition for inhalation can be mediated by a dosing timing element which can include a timer, a dose counter, time measuring device, or a time indicator which when incorporated into the device enables dose tracking, compliance monitoring, and/or dose triggering to a patient during administration of the aerosol medicament.

The term "powder" means a composition that consists of finely dispersed solid particles that are free flowing and capable of being readily dispersed in an inhalation device and subsequently inhaled by a subject so that the particles reach the lungs to permit penetration into the alveoli. Thus, the powder is said to be "respirable." Preferably the average particle size is less than about 10 μιη in diameter preferably with a relatively uniform spheroidal shape distribution. More preferably the diameter is less than about 7.5 μ m and most preferably less than about 5.0 μ m. Usually the particle size distribution is between about 0.1 μ m and about 5 μ m in diameter, particularly about 0.3 μ m to about 5 μ m.

The term "dry" means that the composition has a moisture content below about 10% by weight (% w) water, usually below about 5% w and preferably less it than about 3% w. A dry composition can be such that the particles are readily dispersible in an inhalation device to form an aerosol.

The types of pharmaceutical excipients that are useful as carrier include stabilizers such as human serum albumin (HSA), bulking agents such as carbohydrates, amino acids and polypeptides; pH adjusters or buffers; salts such as sodium chloride; and the like. These carriers may be in a crystalline or amorphous form or may be a mixture of the two.

Suitable pH adjusters or buffers include organic salts prepared from organic acids and bases, such as sodium citrate, sodium ascorbate, and the like; sodium citrate is preferred. Pulmonary administration of a micellar single stranded oligonucleotide formulation may be achieved through metered dose spray devices with propellants such as tetrafluoroethane, heptafluoroethane, dimethylfluoropropane, tetrafluoropropane, butane, isobutane, dimethyl ether and other non-CFC and CFC propellants.

Exemplary devices include devices which are introduced into the vasculature, e.g. , devices inserted into the lumen of a vascular tissue, or which devices themselves form a part of the vasculature, including stents, catheters, heart valves, and other vascular devices. These devices, e.g. , catheters or stents, can be placed in the vasculature of the lung, heart, or leg.

Other devices include non- vascular devices, e.g. , devices implanted in the

peritoneum, or in organ or glandular tissue, e.g. , artificial organs. The device can release a therapeutic substance in addition to a single stranded oligonucleotide, e.g. , a device can release insulin.

In one embodiment, unit doses or measured doses of a composition that includes single stranded oligonucleotide are dispensed by an implanted device. The device can include a sensor that monitors a parameter within a subject. For example, the device can include pump, e.g. , and, optionally, associated electronics.

Tissue, e.g. , cells or organs can be treated with a single stranded oligonucleotide, ex vivo and then administered or implanted in a subject. The tissue can be autologous, allogeneic, or xenogeneic tissue. E.g. , tissue can be treated to reduce graft v. host disease . In other embodiments, the tissue is allogeneic and the tissue is treated to treat a disorder characterized by unwanted gene expression in that tissue. E.g. , tissue, e.g. , hematopoietic cells, e.g. , bone marrow hematopoietic cells, can be treated to inhibit unwanted cell proliferation. Introduction of treated tissue, whether autologous or transplant, can be combined with other therapies. In some implementations, the single stranded oligonucleotide treated cells are insulated from other cells, e.g. , by a semi-permeable porous barrier that prevents the cells from leaving the implant, but enables molecules from the body to reach the cells and molecules produced by the cells to enter the body. In one embodiment, the porous barrier is formed from alginate.

In one embodiment, a contraceptive device is coated with or contains a single stranded oligonucleotide. Exemplary devices include condoms, diaphragms, IUD

(implantable uterine devices, sponges, vaginal sheaths, and birth control devices. Dosage

In one aspect, the invention features a method of administering a single stranded oligonucleotide {e.g., as a compound or as a component of a composition) to a subject {e.g., a human subject). In some embodimetns, the methods involve administering a compound (e.g., a compound of the general formula A-B-C, as disclosed herein, or an single stranded oligonucleotide,) in a unit dose to a subject. In one embodiment, the unit dose is between about 10 mg and 25 mg per kg of bodyweight. In one embodiment, the unit dose is between about 1 mg and 100 mg per kg of bodyweight. In one embodiment, the unit dose is between about 0.1 mg and 500 mg per kg of bodyweight. In some embodiments, the unit dose is more than 0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1, 2, 5, 10, 25, 50 or 100 mg per kg of bodyweight.

The defined amount can be an amount effective to treat or prevent a disease or disorder, e.g., a disease or disorder associated with the SMN1 or SMN2. The unit dose, for example, can be administered by injection {e.g., intravenous or intramuscular), an inhaled dose, or a topical application.

In some embodiments, the unit dose is administered daily. In some embodiments, less frequently than once a day, e.g., less than every 2, 4, 8 or 30 days. In another embodiment, the unit dose is not administered with a frequency {e.g., not a regular frequency). For example, the unit dose may be administered a single time. In some embodiments, the unit dose is administered more than once a day, e.g., once an hour, two hours, four hours, eight hours, twelve hours, etc.

In one embodiment, a subject is administered an initial dose and one or more maintenance doses of a single stranded oligonucleotide. The maintenance dose or doses are generally lower than the initial dose, e.g., one-half less of the initial dose. A maintenance regimen can include treating the subject with a dose or doses ranging from 0.0001 to 100 mg/kg of body weight per day, e.g., 100, 10, 1, 0.1, 0.01, 0.001, or 0.0001 mg per kg of bodyweight per day. The maintenance doses may be administered no more than once every 1, 5, 10, or 30 days. Further, the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease, its severity and the overall condition of the patient. In some embodiments the dosage may be delivered no more than once per day, e.g., no more than once per 24, 36, 48, or more hours, e.g., no more than once for every 5 or 8 days. Following treatment, the patient can be monitored for changes in his condition and for alleviation of the symptoms of the disease state. The dosage of the oligonucleotide may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disease state is observed, if the disease state has been ablated, or if undesired side-effects are observed.

The effective dose can be administered in a single dose or in two or more doses, as desired or considered appropriate under the specific circumstances. If desired to facilitate repeated or frequent infusions, implantation of a delivery device, e.g. , a pump, semipermanent stent (e.g. , intravenous, intraperitoneal, intracisternal or intracapsular), or reservoir may be advisable.

In some embodiments, the pharmaceutical composition includes a plurality of single stranded oligonucleotide species. In some embodiments, the pharmaceutical composition comprises a first single stranded oligonucleotide complementary with a PRC2-associated region of a gene (e.g., SMN1 or SMN2), and a second single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of a gene (e.g., SMN1 or SMN2). In some embodiments, the pharmaceutical composition includes a compound comprising the general formula A-B-C, in which A is a single stranded oligonucleotide complementary with a PRC2-associated region of a gene, B is a linker, and C is a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene.

In another embodiment, the single stranded oligonucleotide species has sequences that are non-overlapping and non-adjacent to another species with respect to a naturally occurring target sequence (e.g. , a PRC2-associated region). In another embodiment, the plurality of single stranded oligonucleotide species is specific for different PRC2-associated regions. In another embodiment, the single stranded oligonucleotide is allele specific. In some cases, a patient is treated with a single stranded oligonucleotide in conjunction with other therapeutic modalities.

Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the compound of the invention is administered in maintenance doses, ranging from 0.0001 mg to 100 mg per kg of body weight.

The concentration of the single stranded oligonucleotide composition is an amount sufficient to be effective in treating or preventing a disorder or to regulate a physiological condition in humans. The concentration or amount of single stranded oligonucleotide administered will depend on the parameters determined for the agent and the method of administration, e.g. nasal, buccal, pulmonary. For example, nasal formulations may tend to require much lower concentrations of some ingredients in order to avoid irritation or burning of the nasal passages. It is sometimes desirable to dilute an oral formulation up to 10- 100 times in order to provide a suitable nasal formulation.

Certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a single stranded oligonucleotide can include a single treatment or, preferably, can include a series of treatments. It will also be appreciated that the effective dosage of a single stranded oligonucleotide used for treatment may increase or decrease over the course of a particular treatment. For example, the subject can be monitored after administering a single stranded oligonucleotide composition. Based on information from the monitoring, an additional amount of the single stranded

oligonucleotide composition can be administered.

Dosing is dependent on severity and responsiveness of the disease condition to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of disease state is achieved. Optimal dosing schedules can be calculated from measurements of SMN1 or SMN2 expression levels in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing

methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual compounds, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In some embodiments, the animal models include transgenic animals that express a human SMN1 or SMN2. In another embodiment, the composition for testing includes a single stranded oligonucleotide that is complementary, at least in an internal region, to a sequence that is conserved between SMN1 or SMN2 in the animal model and the SMN1 or SMN2 in a human.

In one embodiment, the administration of the single stranded oligonucleotide composition is parenteral, e.g. intravenous (e.g. , as a bolus or as a diffusible infusion), intradermal, intraperitoneal, intramuscular, intrathecal, intraventricular, intracranial, subcutaneous, transmucosal, buccal, sublingual, endoscopic, rectal, oral, vaginal, topical, pulmonary, intranasal, urethral or ocular. Administration can be provided by the subject or by another person, e.g., a health care provider. The composition can be provided in measured doses or in a dispenser which delivers a metered dose. Selected modes of delivery are discussed in more detail below.

Kits

In certain aspects of the invention, kits are provided, comprising a container housing a composition comprising a single stranded oligonucleotide. In some embodiments, the kits comprise a container housing a single stranded oligonucleotide complementary with of a PRC2-associated region of a gene; and a second container housing a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, the kits comprise a container housing a single stranded oligonucleotide complementary with of a PRC2- associated region of a gene and a single stranded oligonucleotide complementary to a splice control sequence of a precursor mRNA of the gene. In some embodiments, the composition is a pharmaceutical composition comprising a single stranded oligonucleotide and a pharmaceutically acceptable carrier. In some embodiments, the individual components of the pharmaceutical composition may be provided in one container. Alternatively, it may be desirable to provide the components of the pharmaceutical composition separately in two or more containers, e.g., one container for single stranded oligonucleotides, and at least another for a carrier compound. The kit may be packaged in a number of different configurations such as one or more containers in a single box. The different components can be combined, e.g., according to instructions provided with the kit. The components can be combined according to a method described herein, e.g., to prepare and administer a pharmaceutical composition. The kit can also include a delivery device.

The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference. EXAMPLES

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.

Example 1: Oligonucleotides targeting PRC2-associated regions that upregulate SMN1.

MATERIALS AND METHODS: Real Time PCR

RNA was harvested from the cells using Promega SV 96 Total RNA Isolation system or Trizol omitting the DNAse step. In separate pilot experiments, 50 ng of RNA was determined to be sufficient template for the reverse transcriptase reaction. RNA harvested from cells was normalized so that 50ng of RNA was input to each reverse transcription reaction. For the few samples that were too dilute to reach this limit, the maximum input volume was added. Reverse transcriptase reaction was performed using the Superscript II kit and real time PCR performed on cDNA samples using icycler SYBR green chemistry

(Biorad). A baseline level of mRNA expression for each target gene was determined through quantitative PCR as outlined above. Baseline levels were also determined for mRNA of various housekeeping genes which are constitutively expressed. A "control" housekeeping gene with approximately the same level of baseline expression as the target gene was chosen for comparison purposes.

Protein expression (ELISA)

ELISA to determine SMN protein was carried out per manufacturer's instructions (SMN ELISA kit #ADI-900-209, Enzo Life Sciences). Data was normalized to total protein as measured by bicinchoninic acid (BCA) assay (Pierce cat#: 23225).

Cell Culture

Human hepatocyte Hep3B, human hepatocyte HepG2 cells, mouse hepatoma Hepal-6 cells, and human renal proximal tubule epithelial cells (RPTEC) were cultured using conditions known in the art (see, e.g. Current Protocols in Cell Biology). Other cell lines tested were neuronal cell lines (SK-N-AS, U-87) and SMN patient fibroblasts. Details of the cell lines used in the experiments described herein are provided in Table 5.

Table 5. Cell lines

Oligonucleotide design

Oligonucleotides were designed within PRC2-interacting regions in order to upregulate SMNl. The sequence and structure of each oligonucleotide is shown in Table 2. The following table provides a description of the nucleotide analogs, modifications and intranucleotide linkages used for certain oligonucleotides tested and described in Table 2. In vitro transfection of cells with oligonucleotides

Cells were seeded into each well of 24- well plates at a density of 25,000 cells per 500uL and transfections were performed with Lipofectamine and the single stranded oligonucleotides. Control wells contained Lipofectamine alone. At 48 hours post- transfection, approximately 200 uL of cell culture supernatants were stored at -80 C for ELISA. At 48 hours post-transfection, RNA was harvested from the cells and quantitative PCR was carried out as outlined above. The percent induction of target mRNA expression by each oligonucleotide was determined by normalizing mRNA levels in the presence of the oligonucleotide to the mRNA levels in the presence of control (Lipofectamine alone). This was compared side-by-side with the increase in mRNA expression of the "control" housekeeping gene.

RESULTS:

In vitro delivery of single stranded oligonucleotides upregulated SMNl expression

Oligonucleotides were designed as candidates for upregulating SMNl expression. A total of 52 single stranded oligonucleotides were designed to be complementary to a PRC2- interacting region within a sequence as set forth in SEQ ID NO: 1, 2, 4, or 5.

Oligonucleotides were tested in at least duplicate. The sequence and structural features of the oligonucleotides are set forth in Table 2. Briefly, cells were transfected in vitro with the oligonucleotides as described above. SMNl expression in cells following treatment was evaluated by qRT-PCR. Oligonucleotides that upregulated SMNl expression were identified. Further details are outlined in Table 2.

Table 7 shows further results from experiments in which oligonucleotides were transfected into cells at a particular concentration [oligo] and 48 or 72h later RNA was prepared and qRTPCR assays carried out to determine mRNA levels of full length (FL) or delta7 SMN. In other cases, oligos were administered gymnotically to cells at 10μΜ and RNA harvested 9 days post treatment. The cell lines tested were neuronal cell lines (SK-N- AS, U-87) and SMN patient fibroblasts.

Table 8 shows results from experiments in which oligonucleotides were transfected into cells in combination with either one or two more oligos or small molecule compounds at a particular concentration ([oligo], [2nd], [3rd]) and 48 or 72h later RNA was prepared and qRTPCR assays carried out to determine mRNA levels of full length (FL) or delta7 SMN. The cell lines tested were SMN patient fibroblasts.

Table 9 shows results from experiments in which oligonucleotides were transfected into cells in combination with either one or two more oligos or as dimers or by gymnotic treatment at a particular concentration ([oligo], [2nd], [3rd]) and 24, 48, 72 or 216h later cell lysates were prepared and ELISA assays carried out to determine SMN protein levels. The cell lines tested were SMN patient fibroblasts.

Tables Table 1: Non-Seed hexamer sequences.

AAAAAA, AAAAAG, AAAACA, AAAAGA, AAAAGC, AAAAGG, AAAAUA, AAACAA, AAACAC, AAACAG, AAACAU, AAACCC, AAACCU, AAACGA, AAACGC, AAACGU, AAACUA, AAACUC, AAACUU, AAAGAU, AAAGCC, AAAGGA, AAAGGG, AAAGUC, AAAUAC, AAAUAU, AAAUCG, AAAUCU, AAAUGC, AAAUGU, AAAUUA, AAAUUG, AACAAC, AACAAG, AACAAU, AACACA, AACACG, AACAGA, AACAGC, AACAGG, AACAUC, AACAUG, AACCAA, AACCAC, AACCAG, AACCAU, AACCCC, AACCCG, AACCGA, AACCGC, AACCGG, AACCUA, AACCUU, AACGAA, AACGAC, AACGAG, AACGAU, AACGCU, AACGGG, AACGGU, AACGUA, AACGUC, AACGUG, AACGUU, AACUAU, AACUCA, AACUCC, AACUCG, AACUGA, AACUGC, AACUGU, AACU UA, AACUUC, AACU UG, AACUUU, AAGAAA, AAGAAG, AAGAAU, AAGACG, AAGAGA, AAGAGC, AAGAGG, AAGAGU, AAGAUU, AAGCAA, AAGCAC, AAGCAG, AAGCAU, AAGCCA, AAGCCC, AAGCCG, AAGCCU, AAGCGA, AAGCGG, AAGCGU, AAGCUA, AAGGAA, AAGGAC, AAGGCU, AAGGGC, AAGGGU, AAGGUU, AAGUAA, AAGUAC, AAGUAU, AAGUCC, AAGUCG, AAGUGA, AAGUGG, AAGUUA, AAGU UU, AAUAAA, AAUAAC, AAUAAG, AAUAAU, AAUACA, AAUACC, AAUACG, AAUAGA, AAUAGC, AAUAGG, AAUAGU, AAUAUC, AAUAU U, AAUCAA, AAUCAU, AAUCCA, AAUCCC, AAUCCG, AAUCGA, AAUCGC, AAUCGU, AAUCUA, AAUCUG, AAUCUU, AAUGAA, AAUGAC, AAUGAG, AAUGAU, AAUGCG, AAUGCU, AAUGGA, AAUGGU, AAUGUA, AAUGUC, AAUGUG, AAUUAA, AAUUAC, AAUUAG, AAU UCC, AAU UCG, AAUUGA, AAUUGG, AAU UGU, AAUUUC, AAU UUG, ACAAAA, ACAAAC, ACAAAG, ACAAAU, ACAACC, ACAACG, ACAACU, ACAAGA, ACAAGC, ACAAGU, ACAAUC, ACAAUG, ACAAUU, ACACAG, ACACCA, ACACCC, ACACCG, ACACCU, ACACGA, ACACGC, ACACGU, ACACUC, ACACUG, ACACUU, ACAGAA, ACAGAC, ACAGCC, ACAGCG, ACAGCU, ACAGGG, ACAG UC, ACAGUG, ACAGU U, ACAUAA, ACAUAC, ACAUCC, ACAUCG, ACAUCU, ACAUGA, ACAUGC, ACAUGU, ACAU UG, ACAU UU, ACCAAA, ACCAAC, ACCAAG, ACCAAU, ACCACC, ACCACG, ACCAGA, ACCAGU, ACCAUA, ACCAUG, ACCAU U, ACCCAA, ACCCAC, ACCCCA, ACCCCG, ACCCGA, ACCCGC, ACCCUA, ACCCUC, ACCCUU, ACCGAA, ACCGAC, ACCGAU, ACCGCA, ACCGCC, ACCGCG, ACCGCU, ACCGGA, ACCGGC, ACCGGU, ACCGUA, ACCGUC, ACCGUG, ACCGUU, ACCUAA, ACCUAC, ACCUAG, ACCUAU, ACCUCA, ACCUCC, ACCUCG, ACCUCU, ACCUGA, ACCUGC, ACCUGU, ACCUUA, ACCUUC, ACCU U U, ACGAAA, ACGAAC, ACGAAG, ACGAAU, ACGACA, ACGACC, ACGACG, ACGACU, ACGAGA, ACGAGC, ACGAGG, ACGAGU, ACGAUA, ACGAUC, ACGAUG, ACGAUU, ACGCAA, ACGCAG, ACGCAU, ACGCCC, ACGCCG, ACGCCU, ACGCGA, ACGCGG, ACGCGU, ACGCUA, ACGCUG, ACGCUU, ACGGAA, ACGGAC, ACGGAG, ACGGAU, ACGGCC, ACGGCG, ACGGCU, ACGGGC, ACGGGG, ACGGGU, ACGGUA, ACGGUC, ACGGUG, ACGGUU, ACGUAA, ACGUAC, ACGUAU, ACGUCC, ACGUCG, ACGUCU, ACGUGA, ACGUGC, ACGUGG, ACGUGU, ACGUUA, ACGU UC, ACGUUG, ACGUU U, AC U AAA, ACUAAG, ACUAAU, ACUACA, ACUACC, ACUACG, ACUACU, ACUAGG, ACUAUC, ACUAUG, ACUAUU, ACUCAU, ACUCCC, ACUCCG, ACUCCU, ACUCGA, ACUCGC, ACUCGG, ACUCUC, ACUCUU, ACUGAG, ACUGAU, ACUGCC, ACUGCG, ACUGCU, ACUGGG, ACUGGU, ACUGUC, ACUUAA, ACU UAC, ACUUAU, ACU UCA, ACUUCC, ACUUCG, ACUUCU, ACUUGA, ACU UGC, ACU UGU, ACUUUA, ACU UUC, ACUU UG, AGAAAA, AGAAAC, AGAAAG, AGAACC, AGAACG, AGAACU, AGAAGC, AGAAGU, AGAAUA, AGAAUC, AGAAUG, AGAAUU, AGACAA, AGACAC, AGACAU, AGACCA, AGACCC, AGACCG, AGACCU, AGACGA, AGACGC, AGACGU, AGACUA, AGACUC, AGACUU, AGAGAC,

AGAGAG, AGAGAU, AGAGCC, AGAGCG, AGAGCU, AGAGGC, AGAGGG, AGAGGU, AGAGUA, AGAGUU, AGAUAC, AGAUAG, AGAUAU, AGAUCC, AGAUCG, AGAUCU, AGAUGA, AGAUGC, AGAUGG, AGAU UA, AGAU UC, AGAUUG, AGAUU U, AGCAAC, AGCACA, AGCACG, AGCACU, AGCAGA, AGCAUA, AGCAUC, AGCAUG, AGCCAA, AGCCAU, AGCCCA, AGCCGA, AGCCGC, AGCCGG, AGCCGU, AGCCUA, AGCCUC, AGCGAA, AGCGAG, AGCGAU, AGCGCA, AGCGCC, AGCGCG, AGCGCU, AGCGGA, AGCGGC, AGCGGU, AGCGUA, AGCGUC, AGCGUG, AGCGUU, AGCUAA, AGCUAC, AGCUAG, AGCUAU, AGCUCA, AGCUCC, AGCUCG, AGCUCU, AGCUGA, AGCUGG, AGCUGU, AGCU UC, AGCUU U, AGGAAU, AGGACC, AGGACG, AGGAGA, AGGAGU, AGGAUA, AGGCAA, AGGCAU, AGGCCG, AGGCGA, AGGCGC, AGGCGG, AGGCUA, AGGCUC, AGGCUU, AGGGAC, AGGGAU, AGGGGA, AGGGGU, AGGGUA, AGGGUG, AGGUAA, AGGUAC, AGGUCA, AGGUCC, AGGUCU, AGGUGA, AGGUGC, AGGUGG, AGGUGU, AGGU UC,

AGGUUG, AGUAAA, AGUAAG, AGUAAU, AGUACA, AGUACG, AGUAGC, AGUAGG, AGUAUA, AGUAUC, AGUAUG, AGUAUU, AGUCAA, AGUCAC, AGUCAG, AGUCAU, AGUCCA, AGUCCG, AGUCCU, AGUCGA, AGUCGC, AGUCGG, AGUCGU, AGUCUA, AGUCUC, AGUCUG, AGUCU U, AGUGAA, AGUGAC, AGUGCG, AGUGGG, AGUGUC, AGU UAA, AGU UAC, AGUUAG, AGUUCC, AGU UCG, AGUUGA, AGUUGC,

AGUUGU, AGUUUA, AGUUUC, AGUU UG, AGU UU U, AUAAAC, AUAAAU, AUAACA, AUAACC, AUAACG, AUAACU, AUAAGA, AUAAGC, AUAAGG, AUAAGU, AUAAUC, AUAAUG, AUAAUU, AUACAC, AUACAG, AUACAU, AUACCA, AUACCC, AUACCG, AUACGA, AUACGC, AUACGG, AUACGU, AUACUA, AUACUC, AUACUG, AUACUU, AUAGAA, AUAGAC, AUAGAU, AUAGCA, AUAGCG, AUAGCU, AUAGGA, AUAGGU, AUAGUA, AUAGUC, AUAGUG, AUAGUU, AUAUAC, AUAUAG, AUAUCC, AUAUCG, AUAUCU, AUAUGA, AUAUGC, AUAUGG, AUAUGU, AUAUUC, AUAU UG, AUAUU U, AUCAAA, AUCAAC, AUCAAG, AUCAAU, AUCACA, AUCACC, AUCACG, AUCAGC, AUCAGG, AUCCAA, AUCCAU, AUCCCC, AUCCCG, AUCCGA, AUCCGC, AUCCGG, AUCCUA, AUCCUC, AUCCUG, AUCGAA, AUCGAC, AUCGAG, AUCGAU, AUCGCA, AUCGCC, AUCGCG, AUCGCU, AUCGGC, AUCGGG, AUCGGU, AUCGUC, AUCGUG, AUCGU U, AUCUAA, AUCUAC, AUCUAG, AUCUAU, AUCUCC, AUCUCG, AUCUGU, AUCUUG, AUCUU U, AUGAAA, AUGAAC, AUGAAG, AUGAAU, AUGACC, AUGACU, AUGAGG, AUGAGU, AUGAUA, AUGAUC, AUGAU U, AUGCAA, AUGCAG, AUGCCA, AUGCCC, AUGCCG, AUGCGA, AUGCGG, AUGCGU, AUGCUC, AUGCUU, AUGGAC, AUGGCC, AUGGGA, AUGGGC, AUGGGU, AUGGUC, AUGGUG, AUGUAC, AUGUAU, AUGUCA, AUGUCC, AUGUCG, AUGUGU, AUGUUA, AUGUUC, AU UAAA, AUUAAC, AU UAAG, AU UAAU, AU UACA, AUUACC, AUUACG, AUUACU, AU UAGA, AUUAGC, AUUAGG, AUUAGU, AU UAUA, AUUAUC, AUUAUG, AUUCAC, AUUCCA, AU UCCG, AU UCCU, AUUCGA, AUUCGC, AUUCGG, AU UCGU, AUUCUA, AUUCUC, AUUCU U, AUUGAA, AUUGAC, AUUGAU, AU UGCC, AUUGCG, AU UGCU, AUUGGA, AUUGGC,

AUUGGG, AU UGGU, AUUGUA, AUUGUC, AUUGUG, AU UGU U, AU UUAA, AU UUAG, AU UUAU, AUU UCC, AUU UCG, AUU UCU, AU UUGA, AU UUGC, AU UUGU, AU UUUA, AU UU UC, AUU UUG, AUU UU U, CAAAAG, CAAACA, CAAACC, CAAACG, CAAACU, CAAAGA, CAAAGG, CAAAUA, CAAAUU, CAACAC, CAACAU, CAACCA, CAACCC, CAACCG, CAACGA, CAACGC, CAACGG, CAACGU, CAACUA, CAACUC, CAACUG, CAACUU, CAAGAA, CAAGAC, CAAGAU, CAAGCA, CAAGCC, CAAGCG, CAAGCU, CAAGGA, CAAGGG, CAAGUC, CAAGUG, CAAGU U, CAAUAA, CAAUAC, CAAUAG, CAAUCC, CAAUCG, CAAUCU, CAAUGA, CAAUGC, CAAUGG, CAAUGU, CAAU UC, CAAU UG, CAAU UU, CACAAU, CACACA, CACACG, CACACU, CACAGA, CACAGC, CACAGG, CACAUA, CACAUC, CACAUU, CACCAA, CACCAC, CACCAU, CACCCA, CACCCC, CACCCG, CACCGA, CACCGC, CACCGG, CACCGU, CACCUA, CACCU U, CACGAA, CACGAC, CACGAG, CACGAU, CACGCA, CACGCC, CACGCU, CACGGA, CACGGC, CACGGG, CACGG U, CACGUA, CACGUC, CACGUG, CACGU U, CACUAA, CACUAG, CACUAU, CACUCA, CACUCG, CACUGA, CACUGC, CACUGG, CACUUA, CACU UC, CACU UU, CAGAAA, CAGAAG, CAGAAU, CAGACC, CAGACG, CAGAGC, CAGAUA, CAGAUC, CAGCCG, CAGCCU, CAGCGA, CAGCGC, CAGCGG, CAGCGU, CAGCUC, CAGCUU, CAGGAU, CAGGGG, CAGGGU, CAGGUA, CAGGUC, CAGGUU, CAGUAC, CAGUCG, CAGU UG, CAUAAA, CAUAAC, CAUAAG, CAUAAU, CAUACA, CAUACC, CAUACG, CAUACU, CAUAGA, CAUAGG, CAUAGU, CAUAUA, CAUAUC, CAUAUG, CAUCAA, CAUCAC, CAUCAG, CAUCAU, CAUCCA, CAUCCC, CAUCCG, CAUCGA, CAUCGC, CAUCGG, CAUCGU, CAUCUA, CAUCUC, CAUCUG, CAUCUU, CAUGAA, CAUGAC, CAUGAG, CAUGAU, CAUGCA, CAUGCC, CAUGCG, CAUGCU, CAUGGC, CAUGGG, CAUGGU, CAUGUA, CAUGUC, CAUGUU, CAU UAA, CAUUAC, CAUUAG, CAUUCA, CAU UCC, CAU UCG, CAU UCU, CAU UGA, CAU UGG, CAUU UC, CAU UUG, CAUU UU, CCAAAA, CCAAAC, CCAAAG, CCAAAU, CCAACA, CCAACC, CCAACG, CCAACU, CCAAGA, CCAAGC, CCAAGG, CCAAUC, CCAAUG, CCAAU U, CCACAA, CCACAC, CCACAG, CCACAU, CCACCA, CCACCC, CCACCG, CCACCU, CCACGA, CCACGC, CCACGG, CCACGU, CCACUA, CCACUC, CCACUU, CCAGAA, CCAGAC, CCAGAG, CCAGCC, CCAGGU, CCAGUC, CCAGUU, CCAUAA, CCAUAC, CCAUAG, CCAUAU, CCAUCA, CCAUCC, CCAUCU, CCAUGA, CCAUGC, CCAUGG, CCAUUC, CCAUUG, CCAUU U, CCCAAC, CCCAAG, CCCAAU, CCCACA, CCCAGA, CCCAGC, CCCAGU, CCCAUA, CCCAUC, CCCAUG, CCCAUU, CCCCAA, CCCCAG, CCCCAU, CCCCCC, CCCCCG, CCCCCU, CCCCGA, CCCCGC, CCCCGU, CCCCUA, CCCCUC, CCCGAA, CCCGAC, CCCGAU, CCCGCA, CCCGCU, CCCGGA, CCCGGC, CCCGUA, CCCGUG, CCCGU U, CCCUAA, CCCUAG, CCCUCA, CCCUCU, CCCUGC, CCCUUA, CCCU UC, CCCU UU, CCGAAA, CCGAAC, CCGAAU, CCGACA, CCGACC, CCGACG, CCGACU, CCGAGA, CCGAGG, CCGAGU, CCGAUA, CCGAUC, CCGAUG, CCGAU U, CCGCAA, CCGCAC, CCGCAG, CCGCAU, CCGCCA, CCGCCC, CCGCCG, CCGCCU, CCGCGA, CCGCGC, CCGCGG, CCGCGU, CCGCUA, CCGCUC, CCGCUG, CCGCU U, CCGGAA, CCGGAU, CCGGCA, CCGGCC, CCGGCG, CCGGCU, CCGGGA, CCGGGC, CCGGGG, CCGGGU, CCGGUA, CCGGUC, CCGGUG, CCGUAA, CCGUAG, CCGUAU, CCGUCA, CCGUCC, CCGUCG, CCGUGA, CCGUGU, CCGUUA, CCGUUC, CCGU UG, CCGUU U, CCUAAC, CCUAAG, CCUAAU, CCUACA, CCUACC, CCUACG, CCUACU, CCUAGA, CCUAGC, CCUAGG, CCUAGU, CCUAUA, CCUAUC, CCUAUG, CCUAUU, CCUCAA, CCUCAC, CCUCAG, CCUCAU, CCUCCA, CCUCCC, CCUCCG, CCUCGA, CCUCGC, CCUCGG, CCUCGU, CCUCUA, CCUCUG, CCUGAC, CCUGAU, CCUGCA, CCUGGG, CCUGGU, CCUGU U, CCUUAA, CCUUAC, CCUUAG, CCUUAU, CCUUCG, CCUUGA, CCUUGU, CCU UUA, CCUU UC, CCU UU U, CGAAAA, CGAAAC, CGAAAG, CGAAAU, CGAACA, CGAACC, CGAACG, CGAACU, CGAAGA, CGAAGC, CGAAGG, CGAAGU, CGAAUA, CGAAUC, CGAAUG, CGAAUU, CGACAA, CGACAC, CGACAU, CGACCA, CGACCU, CGACGA, CGACGC, CGACGG, CGACGU, CGACUA, CGACUG, CGACU U, CGAGAA, CGAGAC, CGAGAG, CGAGAU, CGAGCA, CGAGCC, CGAGCG, CGAGCU, CGAGGC, CGAGGG, CGAGGU, CGAGUA, CGAGUC, CGAGUG, CGAGUU, CGAUAA, CGAUAC, CGAUAG, CGAUAU, CGAUCA, CGAUCC, CGAUCG, CGAUCU, CGAUGA, CGAUGC, CGAUGG, CGAUGU, CGAUUA, CGAU UC, CGAUUG, CGAUU U, CGCAAA, CGCAAC, CGCAAG, CGCAAU, CGCACA, CGCACC, CGCACG, CGCAGA, CGCAGC, CGCAGG, CGCAGU, CGCAUA, CGCAUC, CGCAUG, CGCAU U, CGCCAA, CGCCAC, CGCCAG, CGCCAU, CGCCCA, CGCCCC, CGCCCG, CGCCGA, CGCCGC, CGCCGG, CGCCGU, CGCCUA, CGCCUG, CGCCUU, CGCGAA, CGCGAC, CGCGAG, CGCGAU, CGCGCA, CGCGCC, CGCGCG, CGCGCU, CGCGGA, CGCGGC, CGCGGG, CGCGGU, CGCGUA, CGCGUC, CGCGUG, CGCGU U, CGCUAA, CGCUAC, CGCUAG, CGCUAU, CGCUCA, CGCUCC, CGCUCG, CGCUCU, CGCUGA, CGCUGC, CGCUGG, CGCUGU, CGCUUA, CGCU UC, CGCU UG, CGGAAA, CGGAAC, CGGAAG, CGGACA, CGGACC, CGGACG, CGGACU, CGGAGC, CGGAGG, CGGAGU, CGGAUA, CGGAU U, CGGCAA, CGGCAC, CGGCAG, CGGCCA, CGGCCC, CGGCCG, CGGCGC, CGGCGG, CGGCGU, CGGCUA, CGGCUC, CGGCUG, CGGCU U, CGGGAA, CGGGAC, CGGGAG, CGGGAU, CGGGCA, CGGGCC, CGGGCG, CGGGCU, CGGGGU, CGGGUA, CGGGUC, CGGGUG, CGGUAA, CGGUAC, CGGUAG, CGGUAU, CGGUCA, CGGUCG, CGGUCU, CGGUGA, CGGUGG, CGGUGU, CGGU UA, CGGU UC, CGGUUG, CGGUUU, CGUAAA, CGUAAC, CGUAAG, CGUAAU, CGUACA, CGUACG, CGUACU, CGUAGA, CGUAGC, CGUAGG, CGUAGU, CGUAUA, CGUAUC, CGUAUG, CGUAUU, CGUCAA, CGUCAC, CGUCAG, CGUCAU, CGUCCA, CGUCCC, CGUCCG, CGUCCU, CG UCGA, CGUCGG, CGUCGU, CGUCUA, CGUCUC, CGUCUG, CGUCU U, CGUGAA, CGUGAC, CGUGAG, CGUGAU, CGUGCC, CGUGCG, CGUGCU, CGUGGA, CGUGGG, CGUGGU, CGUGUA, CGUGUG, CGUUAA, CGUUAC, CGU UAG,

CGU UAU, CGUUCA, CGUUCC, CGUUCG, CGU UCU, CGUUGA, CGUUGC, CGU UGU, CGU UUA, CGUUUC, CGU U UU, CUAAAA, CUAAAC, CUAAAU, CUAACA, CUAACC, CUAACG, CUAACU, CUAAGA, CUAAGC, CUAAGU, CUAAUA, CUAAUC, CUAAUG, CUACAC, CUACAU, CUACCA, CUACCC, CUACCG, CUACCU, CUACGA, CUACGC, CUACGG, CUACGU, CUACUA, CUACUC, CUACUG, CUAGAA, CUAGAG, CUAGAU, CUAGCA, CUAGCC, CUAGCG, CUAGCU, CUAGGA, CUAGGG, CUAGGU, CUAGUG, CUAGUU, CUAUAA, CUAUAG, CUAUAU, CUAUCA, CUAUCC, CUAUCG, CUAUCU, CUAUGA, CUAUGC, CUAUGG, CUAUGU, CUAUUA, CUAUUG, CUCAAC, CUCAAG, CUCAAU, CUCACC, CUCACG, CUCAGC, CUCAUA, CUCAUC, CUCAUG, CUCAU U, CUCCAC, CUCCCC, CUCCCG, CUCCGA, CUCCGC, CUCCGG, CUCCUA, CUCCUC, CUCCU U, CUCGAA, CUCGAC, CUCGAG, CUCGAU, CUCGCA, CUCGCC, CUCGCG, CUCGGG, CUCGGU, CUCGUA, CUCGUC, CUCGUG, CUCGU U, CUCUAA, CUCUAC, CUCUAU, CUCUCA, CUCUCC, CUCUCU, CUCUGC, CUCUGU, CUCUUA, CUCU UG, CUGAAG, CUGACC, CUGACG, CUGAGC, CUGAUA, CUGAUC, CUGCCG, CUGCCU, CUGCGA, CUGCUA, CUGCUU, CUGGAG, CUGGAU, CUGGCG, CUGGGU, CUGUAC, CUGUCA, CUGUCC, CUGUCG, CUGUGG, CUGUGU, CUGUUA, CUGU UU, CUUAAC, CUUAAG, CU UAAU, CU UACC, CUUACG, CUUAGA, CU UAGC, CU UAGG, CUUAGU, CUUAUA, CU UAUC, CU UAUG, CUUAUU, CU UCAG, CU UCAU, CUUCCA, CUUCCC, CU UCCG, CU UCCU, CUUCGA, CUUCGC, CU UCGG, CUUCGU, CU UCUA, CUUGAC, CUUGAG, CUUGAU, CUUGCA, CUUGCC, CUUGCG, CU UGCU, CUUGGC, CUUGGU, CU UGUU, CUU UAC, CU UUAG, CU UUAU, CUU UCA, CUU UCG, CU UUCU, CUUUGA, CU UUGC, CU UUGU, CU U UUA, CUUU UC, CUU UUG, CUU UUU, GAAAAA, GAAAAG, GAAAAU, GAAACC, GAAACG, GAAAGA, GAAAGC, GAAAGU, GAAAUA, GAAAUC, GAAAUG, GAAAUU, GAACAA, GAACAC, GAACAG, GAACAU, GAACCA, GAACCC, GAACCG, GAACCU, GAACGA, GAACGC, GAACGG, GAACGU, GAACUA, GAACUG, GAACUU, GAAGAC, GAAGAG, GAAGCA, GAAGCG, GAAGCU, GAAGUC, GAAUAA, GAAUAC, GAAUAG, GAAUAU, GAAUCC, GAAUCG, GAAUCU, GAAUGA, GAAUGC, GAAUGU, GAAU UA, GAAU UC, GAAUU U, GACAAA, GACAAG, GACAAU, GACACC, GACAGA, GACAGG, GACAUA, GACAUG, GACAUU, GACCAA, GACCAC, GACCAG, GACCCA, GACCCC, GACCCG, GACCGC, GACCGG, GACCGU, GACCUA, GACCUC, GACCU U, GACGAA, GACGAC, GACGAG, GACGAU, GACGCA, GACGCC, GACGCG, GACGCU, GACGGA, GACGGC, GACGGG, GACGGU, GACGUA, GACGUC, GACGUG, GACGUU, GACUAA, GACUAC, GACUAG, GACUAU, GACUCA, GACUCC, GACUCG, GACUGG, GACUGU, GACUUA, GACUUG, GACUU U, GAGAAU, GAGAGA, GAGAGC, GAGAGG, GAGAUA, GAGAUC, GAGCAA, GAGCAU, GAGCCA, GAGCGA, GAGCGG, GAGCGU, GAGGGU, GAGGUC, GAGGUG, GAGUAA, GAGUAG, GAGUCC, GAGUUC, GAGU UU, GAUAAA, GAUAAC, GAUAAG, GAUAAU, GAUACA, GAUACC, GAUACG, GAUACU, GAUAGA, GAUAGC, GAUAGG, GAUAGU, GAUAUA, GAUCAA, GAUCAC, GAUCAU, GAUCCA, GAUCCC, GAUCCU, GAUCGC, GAUCGG, GAUCGU, GAUCUA, GAUCUG, GAUCU U, GAUGAA, GAUGAC, GAUGAG, GAUGCA, GAUGCC, GAUGCG, GAUGCU, GAUGGC, GAUGGG, GAUGGU, GAUGUG, GAUGUU, GAUUAA, GAUUAC, GAUUAG, GAUUAU, GAUUCA, GAUUCG, GAU UCU, GAUUGA, GAUUGC, GAU UUA, GAU UUC, GAUU UG, GAUUU U, GCAAAC, GCAAAG, GCAAAU, GCAACA, GCAACC, GCAAGC, GCAAGU, GCAAUA, GCAAUC, GCAAUG, GCAAUU, GCACAA, GCACAC, GCACAG, GCACCC, GCACCG, GCACCU, GCACGA, GCACGC, GCACGU, GCACUA, GCACUC, GCACUG, GCACUU, GCAGAU, GCAGCC, GCAGCG, GCAGGC, GCAGUA, GCAGUC, GCAGUG, GCAGUU, GCAUAA, GCAUAG, GCAUAU, GCAUCG, GCAUCU, GCAUGA, GCAUGC, GCAUGG, GCAUGU, GCAU UA, GCAU UC, GCAUUG, GCAUU U, GCCAAA, GCCAAC, GCCAAU, GCCACA, GCCACC, GCCACG, GCCAGA, GCCAGU, GCCAUA, GCCAUC, GCCAUG, GCCAUU, GCCCAA, GCCCAC, GCCCAG, GCCCCG, GCCCGA, GCCCGG, GCCCGU, GCCGAA, GCCGAC, GCCGAG, GCCGAU, GCCGCA, GCCGCU, GCCGGA, GCCGGC, GCCGGG, GCCGGU, GCCGUA, GCCGUC, GCCGUG, GCCGU U, GCCUAA, GCCUAU, GCCUCA, GCCUCC, GCCUCG, GCCUGA, GCCU UA, GCCU UU, GCGAAA, GCGAAC, GCGAAG, GCGAAU, GCGACC, GCGACG, GCGACU, GCGAGA, GCGAGC, GCGAGG, GCGAGU, GCGAUA, GCGAUC, GCGAUG, GCGAUU, GCGCAA, GCGCAC, GCGCAG, GCGCAU, GCGCCA, GCGCCC, GCGCCU, GCGCGA, GCGCGU, GCGCUA, GCGCUC, GCGCUG, GCGCUU, GCGGAA, GCGGAC, GCGGAU, GCGGCA, GCGGCC, GCGGCU, GCGGGA, GCGGUA, GCGGUC, GCGGUU, GCGUAA, GCGUAC, GCGUAG, GCGUAU, GCGUCA, GCGUCC, GCGUCG, GCGUCU, GCGUGA, GCGUGC, GCGUGG, GCGUGU, GCGU UA, GCGUUC, GCGU UG, GCGUU U, GCUAAA, GCUAAC, GCUAAG, GCUAAU, GCUACC, GCUACG, GCUACU, GCUAGA, GCUAGG, GCUAGU, GCUAUA, GCUAUC, GCUAUU, GCUCAA, GCUCAC, GCUCAG, GCUCAU, GCUCCA, GCUCCC, GCUCCG, GCUCGA, GCUCGC, GCUCGU, GCUCUA, GCUCUC, GCUCU U, GCUGAA, GCUGAC, GCUGAU, GCUGCA, GCUGCC, GCUGCG, GCUGCU, GCUGUG, GCUGUU, GCUUAC, GCUUAG, GCUUAU, GCU UCA, GCUUCG, GCU UGA, GCUUGG, GCUUGU, GCUU UA, GCU UUG, GGAAAG, GGAACA, GGAACC, GGAACG, GGAACU, GGAAGU, GGAAUA, GGAAUC, GGAAU U, GGACAA, GGACAC, GGACAG, GGACAU, GGACCG, GGACGA, GGACGC, GGACGU, GGACUA, GGACUC, GGACU U, GGAGAC, GGAGCA, GGAGCG, GGAGGG, GGAGUA, GGAUAA, GGAUAC, GGAUCA, GGAUCC, GGAUCG, GGAUCU, GGAUGC, GGAUUA, GGAUUG, GGCAAU, GGCACA, GGCACU, GGCAGA, GGCAUA, GGCAUC, GGCCAC, GGCCAG, GGCCCC, GGCCGA, GGCCGC, GGCCGU, GGCCUA, GGCCUG, GGCCU U, GGCGAA, GGCGAG, GGCGAU, GGCGCA, GGCGCU, GGCGGU, GGCGUA, GGCGUC, GGCGUG, GGCGU U, GGCUAA, GGCUAC, GGCUAG, GGCUAU, GGCUCC, GGCUCG, GGCUGA, GGCU UA, GGCUUC, GGCUUG, GGGAAU, GGGACA, GGGAGA, GGGAGU, GGGAUA, GGGAU U, GGGCAA, GGGCAC, GGGCAG, GGGCCG, GGGCGG, GGGGCC, GGGGGG,

GGGGGU, GGGGUA, GGGUAC, GGGUAU, GGGUCA, GGGUCC, GGGUCG, GGGUGA, GGGUGC, GGGU UA, GGGU UG, GGUAAA, GGUAAC, GGUAAG, GGUAAU, GGUACA, GGUACC, GGUACG,

GGUACU, GGUAGC, GGUAGG, GGUAGU, GGUAUA, GGUAUC, GGUAUG, GGUCAA, GGUCAC, GGUCAG, GGUCAU, GGUCCA, GGUCCG, GGUCCU, GGUCGA, GGUCGC, GG UCGG, GGUCGU, GGUCUC, GGUCU U, GGUGAA, GGUGAC, GGUGAU, GGUGCA, GGUGCC, GGUGGC, GGUGUA, GGUGUC, GGU UAA, GGU UAG, GGU UAU, GGUUCA, GGU UCC, GGU UCG, GGU UGC, GGU UUC, GGUU UU, GUAAAA, GUAAAG, GUAAAU, GUAACC, GUAACG, GUAACU, GUAAGA, GUAAGC, GUAAGG, GUAAGU, GUAAUA, GUAAUC, GUAAUG, GUAAUU, GUACAA, GUACAC, GUACAG, GUACAU, GUACCA, GUACCC, GUACCG, GUACCU, GUACGA, GUACGC, GUACGG, GUACGU, GUACUA, GUACUC, GUACUG, GUACUU, GUAGAA, GUAGAC, GUAGCA, GUAGCC, GUAGCG, GUAGCU, GUAGGA, GUAGGC, GUAGGG,

GUAGGU, GUAGUA, GUAGUC, GUAUAA, GUAUAC, GUAUAG, GUAUAU, GUAUCA, GUAUCG, GUAUCU, GUAUGA, GUAUGC, GUAUGG, GUAUUA, GUAU UG, G UAU UU, GUCAAA, GUCAAG, GUCAAU, GUCACA, GUCACC, GUCACG, GUCAGA, GUCAGC, GUCAGG, GUCAUA, GUCAUC, GUCAUG, GUCCAA, GUCCAC, GUCCAU, GUCCCC, GUCCCU, GUCCGA, GUCCGC, GUCCGG, GUCCGU, GUCCUA, GUCCUG, GUCCU U, GUCGAA, GUCGAC, GUCGAG, GUCGAU, GUCGCA, GUCGCC, GUCGCG, GUCGCU, GUCGGA, GUCGGC, GUCGGG, GUCGGU, GUCGUA, GUCGUC, GUCGU U, GUCUAA, GUCUAG, GUCUCA, GUCUCC, GUCUCG, GUCUGA, GUCUGG, GUCUGU, GUCU UC, GUCU UU, GUGAAA, GUGAAC, GUGAAG, GUGACC, GUGACG, GUGAGA, GUGAGC, GUGAGU, GUGAUC, GUGAUG, GUGAUU, GUGCAC,

GUGCAU, GUGCCC, GUGCCG, GUGCGA, GUGCGG, GUGCGU, GUGCUA, GUGCUC, GUGCUG,

GUGGAG, GUGGCG, GUGGCU, GUGGGU, GUGGUC, GUGGUG, GUGUAA, GUGUAG, GUGUCG, GUGUGA, GUGUGC, GUGUGU, GUGUUG, GUGU UU, GU UAAA, GUUAAC, GUUAAG, GU UACA, GU UACC, GUUACG, GU UACU, GU UAGA, GUUAGC, GUUAGU, GUUAUA, GUUAUC, GUUAUG, GU UAUU, GUUCAA, GUUCAC, GUUCAG, GU UCCA, GUUCCG, GUUCGA, GU UCGC, GU UCGG, GU UCGU, GU UCUA, GUUCUG, GUUGAA, GU UGAC, GUUGAG, GUUGAU, GUUGCG, GUUGCU, GUUGGA, GU UGGC, GU UGGU, GU UGUC, GUUGUG, GUUGU U, GUU UAA, GU UUAC, GU UUAG, GUU UAU, GU UUCA, GUUUCC, GU UUCU, GU UUGA, GUU UGC, GUU UGG, GUU UGU, GU UUUA, GU UU UC,

GU UU UU, UAAAAA, UAAAAC, UAAAAG, UAAAAU, UAAACA, UAAACC, UAAACG, UAAACU, UAAAGA, UAAAGG, UAAAGU, UAAAUA, UAAAUC, UAAAUG, UAAAU U, U A AC A A, UAACAC, UAACAG, UAACCA, UAACCC, UAACCG, UAACCU, UAACGA, UAACGC, UAACGG, UAACGU, UAACUA, UAACUG, UAACUU, UAAGAG, UAAGAU, UAAGCA, UAAGCC, UAAGCG, UAAGCU, UAAGGA, UAAGGC, UAAGGG, UAAGGU, UAAGUA, UAAGUC, UAAGUG, UAAGUU, UAAUAA, UAAUCA, UAAUCC, UAAUCG, UAAUCU, UAAUGA, UAAUGG, UAAUGU, UAAUUA, UAAU UC, UAAUUG, UACAAC, UACAAG, UACAAU, UACACC, UACACG, UACACU, UACAGA, UACAGC, UACAUA, UACAUC, UACAU U, UACCAA, UACCAC, UACCAG, UACCAU, UACCCC, UACCCG, UACCCU, UACCGA, UACCGC, UACCGG, UACCGU, UACCUA, UACCUG, UACGAA, UACGAC, UACGAG, UACGAU, UACGCA, UACGCC, UACGCG, UACGCU, UACGGC, UACGGG, UACGGU, UACGUA, UACGUC, UACGUG, UACGUU, UACUAA, UACUAC, UACUAG, UACUAU, UACUCA, UACUCC, UACUCG, UACUCU, UACUGA, UACUGC, UACUGG, UACU UA, UACU UG, UACU UU, UAGAAA, UAGAAG, UAGAAU, UAGACA, UAGACG, UAGAGA, UAGAGC, UAGAGU, UAGAUA, UAGAUC, UAGAUG, UAGCAU, UAGCCC, UAGCCG, UAGCCU, UAGCGA, UAGCGC, UAGCGU, UAGCUA, UAGCUC, UAGCUG, UAGGAA, UAGGAU, UAGGCG, UAGGCU, UAGGGU, UAGGUC, UAGGUG, UAGGUU, UAGUAA, UAGUAC, UAGUAG, UAGUAU, UAGUCA, UAGUCG, UAGUGU, UAGUUA, UAGU UC, UAGU UG, UAGUU U,

UAUAAC, UAUAAG, UAUACU, UAUAGA, UAUAGC, UAUAGG, UAUAGU, UAUAUA, UAUAUC, UAUAUG, UAUAU U, UAUCAA, UAUCAC, UAUCAU, UAUCCA, UAUCCC, UAUCCG, UAUCCU, UAUCGA, UAUCGC, UAUCGG, UAUCGU, UAUCUA, UAUCUC, UAUCUG, UAUCUU, UAUGAA, UAUGAC, UAUGAG,

UAUGAU, UAUGCA, UAUGCG, UAUGCU, UAUGGA, UAUGGC, UAUGUC, UAUGUG, UAUGU U, UAU UAG, UAUUCA, UAU UCC, UAUUCG, UAUUCU, UAUUGA, UAUUGG, UAUU UA, UAU UUC,

UAU UUG, UAUUU U, UCAAAA, UCAAAC, UCAAAG, UCAACC, UCAACU, UCAAGA, UCAAGC, UCAAUA, UCAAUC, UCAAUG, UCAAUU, UCACCC, UCACCG, UCACCU, UCACGA, UCACGC, UCACGG, UCACGU, UCACUA, UCACUC, UCACUU, UCAGAA, UCAGAC, UCAGAG, UCAGCG, UCAGCU, UCAGGA, UCAGGC, UCAGGU, UCAGUC, UCAGUU, UCAUAA, UCAUCA, UCAUCC, UCAUCG, UCAUGC, UCAUGG, UCAUGU, UCAUUA, UCAUUG, UCCAAA, UCCAAC, UCCAAG, UCCAAU, UCCACA, UCCACC, UCCACG, UCCAGC, UCCAGG, UCCAUA, UCCAUC, UCCAUU, UCCCAA, UCCCAG, UCCCAU, UCCCCC, UCCCCG, UCCCCU, UCCCGA, UCCCGC, UCCCGG, UCCCGU, UCCCUA, UCCCUC, UCCGAA, UCCGAC, UCCGAG, UCCGAU, UCCGCA, UCCGCC, UCCGGA, UCCGGC, UCCGGU, UCCGUA, UCCGUC, UCCGUG, UCCUAA, UCCUCA, UCCUCG, UCCUCU, UCCUGC, UCCUGU, UCCUUA, UCCUUC, UCCUUU, UCGAAA, UCGAAC, UCGAAG, UCGAAU, UCGACA, UCGACC, UCGACG, UCGACU, UCGAGA, UCGAGC, UCGAGG, UCGAUA, UCGAUC, UCGAUG, UCGAUU, UCGCAA, UCGCAC, UCGCAG, UCGCAU, UCGCCA, UCGCCC, UCGCCG, UCGCCU, UCGCGA, UCGCGC, UCGCGU, UCGCUA, UCGCUC, UCGGAA, UCGGAC, UCGGAG, UCGGAU, UCGGCA, UCGGCU, UCGGGG, UCGGGU, UCGGUC, UCGGUG, UCGGUU, UCGUAA, UCGUAC, UCGUAG,

UCGUAU, UCGUCA, UCGUCC, UCGUCG, UCGUCU, UCGUGA, UCGUGU, UCGUUA, UCGUUC, UCGUUG, UCGUUU, UCUAAC, UCUAAG, UCUAAU, UCUACA, UCUACC, UCUACG, UCUACU, UCUAGC, UCUAGG, UCUAGU, UCUAUA, UCUAUC, UCUAUG, UCUAUU, UCUCAG, UCUCAU, UCUCCG, UCUCGC, UCUCGG, UCUCGU, UCUCUC, UCUGAA, UCUGAU, UCUGCA, UCUGCG, UCUGCU, UCUGGC, UCUGGU, UCUGUC, UCUGUG, UCUGUU, UCUUAA, UCUUAC, UCUUAG, UCUUAU, UCUUCA, UCUUCC, UCUUCG, UCUUCU, UCUUGC, UCUUGG, UCUUGU, UCUUUA, UCUUUC, UCUUUG, UCUUUU, UGAAAA, UGAAAC, UGAACA, UGAACC, UGAAGG, UGAAUC, UGAAUG, UGACAA, UGACAC, UGACAG, UGACCA, UGACCC, UGACCG, UGACGA, UGACGC, UGACGG, UGACGU, UGACUA, UGACUC, UGACUU, UGAGAG, UGAGAU, UGAGCA, UGAGCC, UGAGCU, UGAGGC, UGAGGU, UGAGUA, UGAGUU, UGAUAC, UGAUAG, UGAUAU, UGAUCA, UGAUCG, UGAUCU, UGAUGA, UGAUGC, UGAUGG, UGAUGU, UGAUUA, UGAUUC, UGAUUG, UGAUUU, UGCAAC, UGCAAG, UGCACA, UGCACG, UGCAGG, UGCAGU, UGCAUC, UGCCCA, UGCCCC, UGCCCG, UGCCGA, UGCCGC, UGCCGG, UGCCGU, UGCCUA, UGCCUC, UGCCUG, UGCCUU, UGCGAA, UGCGAC, UGCGAU, UGCGCC, UGCGCG, UGCGCU, UGCGGC, UGCGGG, UGCGGU, UGCGUA, UGCGUC, UGCGUG, UGCGUU, UGCUAC, UGCUAU, UGCUCC, UGCUCG, UGCUGC, UGCUGG, UGCUGU, UGCUUA, UGCUUU, UGGAAC, UGGAAG, UGGAGC, UGGAUC, UGGAUU, UGGCAA, UGGCAC, UGGCAG, UGGCCG, UGGCCU, UGGCGA, UGGCGC, UGGCGU, UGGCUA, UGGCUC, UGGCUU, UGGGAA, UGGGCA, UGGGCC, UGGGGC, UGGGUC, UGGUAA, UGGUAG, UGGUAU, UGGUCC, UGGUCG, UGGUCU, UGGUGA, UGGUGC, UGGUGG, UGGUGU, UGGUUA, UGGUUG, UGUAAA, UGUAAC, UGUAAG, UGUACC, UGUACG, UGUACU, UGUAGA, UGUAGC, UGUAGU, UGUAUC,

UGUAUU, UGUCAA, UGUCAC, UGUCAG, UGUCAU, UGUCCA, UGUCCC, UGUCCG, UGUCGA, UGUCGC, UGUCGG, UGUCGU, UGUCUA, UGUCUC, UGUGAC, UGUGAG, UGUGAU, UGUGCA, UGUGGU, UGUGUA, UGUGUU, UGUUAC, UGUUAG, UGUUAU, UGUUCA, UGUUCC, UGUUCG, UGUUGG, UGUUGU, UGUUUA, UGUUUC, UGUUUG, UGUUUU, UUAAAA, UUAAAC, UUAAAG, UUAAAU, UUAACC, UUAACG, UUAACU, UUAAGU, UUAAUA, UUAAUC, UUAAUG, UUAAUU, UUACAA, UUACAC, UUACAG, UUACAU, UUACCA, UUACCC, UUACCG, UUACCU, UUACGA, UUACGC, UUACGG, UUACGU, UUACUA, UUACUC, UUACUG, UUACUU, UUAGAA, UUAGAC, UUAGCC, UUAGCG, UUAGCU, UUAGGC, UUAGGU, UUAGUA, UUAGUC, UUAGUU, UUAUAA, UUAUAC, UUAUAG, UUAUAU, UUAUCC, UUAUCG, UUAUCU, UUAUGA, UUAUGG, UUAUGU, UUAUUA, UUAUUC, UUAUUG, UUAUUU, UUCAAC, UUCAAU, UUCACA, UUCACC, UUCACG, UUCACU, UUCAGC, UUCAGG, UUCAGU, UUCAUA, UUCAUC, UUCAUG, UUCAUU, UUCCAA, UUCCCA, UUCCCG, UUCCGA, UUCCGU, UUCCUU, UUCGAA, UUCGAC, UUCGAG, UUCGAU, UUCGCA, UUCGCC, UUCGCG, UUCGCU, UUCGGA, UUCGGC, UUCGGG, UUCGGU, UUCGUA, UUCGUC, UUCGUG, UUCGUU, UUCUAC, UUCUAG, UUCUCA, UUCUCG, UUCUGG, UUCUUA, UUCUUU, UUGAAA, UUGAAC, UUGAAG, UUGAAU, UUGACC, UUGACG, UUGACU, UUGAGA, UUGAGC, UUGAGU, UUGAUA, UUGAUC, UUGAUG, UUGAUU, UUGCAA, UUGCAC, UUGCAG, UUGCAU, UUGCCC, UUGCCG, UUGCGA, UUGCGC, UUGCGG, UUGCGU, UUGCUA, 5 UUGCUC, UUGCUG, UUGCUU, UUGGAA, UUGGAG, UUGGCC, UUGGCG, UUGGCU, UUGGGC,

UUGGGU, UUGGUA, UUGGUG, UUGUAA, UUGUAC, UUGUCA, UUGUCG, UUGUCU, UUGUGC, UUGUGG, UUGUUA, UUGUUG, UUGUUU, UUUAAA, UUUAAC, UUUAAG, UUUAAU, UUUACA, UUUACC, UUUACG, UUUACU, UUUAGA, UUUAGC, UUUAGG, UUUAGU, UUUAUA, UUUAUC, UUUAUG, UUUAUU, UUUCAU, UUUCCA, UUUCCG, UUUCCU, UUUCGA, UUUCGC, UUUCGG, 10 UUUCGU, UUUCUA, UUUCUC, UUUCUG, UUUCUU, UUUGAA, UUUGAC, UUUGAG, UUUGAU,

UUUGCC, UUUGCU, UUUGGA, UUUGGC, UUUGGG, UUUGGU, UUUGUA, UUUGUC, UUUGUU, UUUUAA, UUUUAG, UUUUAU, UUUUCC, UUUUCG, UUUUCU, UUUUGA, UUUUGC, UUUUGG, UUUUGU, UUUUUA, UUUUUC, UUUUUU

Table 2: Oligonucleotide sequences made for testing

Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 0.8126719 0.1352513 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21157U20 -01 52 51

SMN1 0.8570321 0.0273187 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21157U20 -01 01 37

SMN1 0.1679989 0.1679986 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21157U20 -01 15 72

SMN1 1.0481253 0.0393027 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21157U20 -01 02 84

SMN1 1.3817042 0.0532905 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21157U20 -01 07 65

SMN1 0.9798692 0.0205152 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21157U20 -01 47 27

SMN1 0.7600003 0.0429932 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21158U20 -02 18 12

SMN1 0.9871384 0.0681879 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21158U20 -02 47 98

SMN1 2.2524945 1.8031906 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21158U20 -02 26 69

SMN1 1.1143879 0.0267332 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21158U20 -02 73 51

SMN1 1.3464192 0.0276412 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21158U20 -02 9 81

SMN1 1.1536970 0.0249999 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21158U20 -02 83 91

SMN1 1.9072297 0.5259392 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21159U20 -03 5 96

SMN1 1.1327582 0.0946401 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21159U20 -03 64 77

SMN1 0.2961917 0.1732823 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21159U20 -03 4 09 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 1.4881793 0.1727195 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21159U20 -03 5 07

SMN1 1.2993282 0.0598252 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21159U20 -03 6 28

SMN1 1.5115678 0.0541781 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21159U20 -03 14 75

SMN1 1.0483065 0.2439345 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21160U20 -04 17 43

SMN1 1.3224072 0.1000223 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21160U20 -04 67 92

SMN1 0.1331700 0.0328243 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21160U20 -04 13 91

SMN1 1.2895501 0.3301959 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21160U20 -04 63 87

SMN1 1.2802254 0.0625779 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21160U20 -04 92 72

SMN1 1.4884827 0.0446412 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21160U20 -04 95 87

SMN1 0.8767475 0.0873925 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21161U20 -05 27 04

SMN1 1.1671203 0.0698140 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21161U20 -05 45 91

SMN1 0.0883178 0.0398870 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21161U20 -05 63 14

SMN1 1.3100532 0.2342313 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21161U20 -05 56 48

SMN1 1.0386996 0.0564213 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21161U20 -05 43 62

SMN1 0.8591447 0.0399700 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21161U20 -05 51 15

SMN1 0.7046598 0.0872441 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21162U20 -06 91 19

SMN1 1.1119400 0.0885713 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21162U20 -06 6 77

SMN1 0.5768596 0.2461865 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21162U20 -06 2 41

SMN1 1.4194188 0.4324471 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21162U20 -06 84 22

SMN1 1.1462517 0.0518915 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21162U20 -06 04 41

SMN1 1.0306823 0.0130708 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21162U20 -06 17 35

SMN1 0.6820857 0.0848853 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21163U20 -07 32 51

SMN1 0.9758535 0.0341785 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21163U20 -07 52 42

SMN1 1.0132523 0.1185407 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21163U20 -07 14 59

SMN1 1.0393819 0.0598153 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21163U20 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 1.1569496 0.1073854 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21163U20 -07 05 05

SMN1 1.2395039 0.1346038 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21163U20 -07 54 44

SMN1 0.9487148 0.1427082 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21164U20 -08 88 31

SMN1 1.3120804 0.0584649 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21164U20 -08 45 93

SMN1 0.2165300 0.1774005 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21164U20 -08 07 55

SMN1 2.0821517 0.8151842 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21164U20 -08 81 52

SMN1 1.0100906 0.2005887 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21164U20 -08 04 91

SMN1 1.2239476 0.2953072 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21164U20 -08 67 43

SMN1 0.7751906 0.0986951 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21165U20 -09 3 18

SMN1 1.6857316 0.0148840 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21165U20 -09 16 28

SMN1 0.6214067 0.2272112 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21165U20 -09 81 61

SMN1 0.8559392 0.2561083 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21165U20 -09 2 37

SMN1 0.9401860 0.1970084 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21165U20 -09 97 64

SMN1 0.8644811 0.1627392 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21165U20 -09 45 71

SMN1 0.9457309 0.0807295 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21166U20 -10 86 2

SMN1 1.5745269 0.1230626 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21166U20 -10 02 84

SMN1 0.4828222 0.1315574 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21166U20 -10 42 74

SMN1 1.2806291 0.1708842 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21166U20 -10 28 5

SMN1 1.1272546 0.1524863 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21166U20 -10 54 74

SMN1 1.0695714 0.1221067 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21166U20 -10 58 58

SMN1 0.7744369 0.0380761 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21167U20 -11 79 82

SMN1 1.5627142 0.1580430 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21167U20 -11 54 98

SMN1 0.4636559 0.2955138 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21167U20 -11 38 86

SMN1 0.9576116 0.3341375 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21167U20 -11 52 41

SMN1 1.2259738 0.2234727 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21167U20 -11 18 58 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 1.0893022 0.1264142 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21167U20 -11 59 68

SMN1 0.9814294 0.0793738 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21168U20 -12 76 4

SMN1 1.5850881 0.0529191 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21168U20 -12 28 2

SMN1 0.2085860 0.1870176 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21168U20 -12 47 55

SMN1 3.2669658 2.0020743 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21168U20 -12 96 69

SMN1 1.0338137 0.2043762 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21168U20 -12 9 91

SMN1 1.1374716 0.2467919 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21168U20 -12 71 54

SMN1 0.7496364 0.1032770 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21169U20 -13 37 03

SMN1 1.1759892 0.1223555 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21169U20 -13 63 85

SMN1 0.1614991 0.0793562 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21169U20 -13 59 87

SMN1 1.2877635 0.0903067 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21169U20 -13 91 17

SMN1 1.3368516 0.1778149 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21169U20 -13 75

SMN1 1.0377722 0.0394045 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21169U20 -13 91 07

SMN1 0.7716351 0.0860419 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21170U20 -14 77 59

SMN1 1.4670485 0.0731138 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21170U20 -14 48 84

SMN1 1.9782541 1.3529511 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21170U20 -14 54 56

SMN1 1.3119909 0.0731216 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21170U20 -14 37 34

SMN1 1.1289277 0.1471627 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21170U20 -14 7 01

SMN1 0.8557951 0.0177971 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21170U20 -14 21 81

SMN1 0.8914919 0.0398220 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21171U20 -15 64 32

SMN1 1.5734403 0.1174530 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21171U20 -15 42 17

SMN1 0.3660431 0.1171620 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21171U20 -15 04 19

SMN1 1.7382173 0.5201481 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21171U20 -15 94 55

SMN1 1.3832013 0.1018307 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21171U20 -15 37 76

SMN1 1.6194950 0.0383649 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21171U20 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 0.7372188 0.0380675 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21172U20 -16 1 83

SMN1 1.4416161 0.0598239 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21172U20 -16 96 44

SMN1 0.5100566 0.2865226 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21172U20 -16 05 59

SMN1 1.3819142 0.2478802 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21172U20 -16 14 29

SMN1 1.3100735 0.0263470 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21172U20 -16 73 93

SMN1 1.4181326 0.0823717 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21172U20 -16 46 08

SMN1 1.2190656 0.2819876 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21173U20 -17 51 74

SMN1 1.2748191 0.1795272 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21173U20 -17 95 93

SMN1 0.4167392 0.0660662 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21173U20 -17 22 42

SMN1 3.3318430 0.9701748 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21173U20 -17 17 73

SMN1 1.2608565 0.0385657 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21173U20 -17 22 99

SMN1 1.6090453 0.1048743 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21173U20 -17 11 4

SMN1 0.8684419 0.0881846 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21174U20 -18 41 98

SMN1 1.2216635 0.0644455 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21174U20 -18 74 39

SMN1 10.284551 3.9293108 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21174U20 -18 67 32

SMN1 1.8009207 0.4255904 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21174U20 -18 64 5

SMN1 1.2617526 0.0698171 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21174U20 -18 02 43

SMN1 1.5927007 0.1992809 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21174U20 -18 96 16

SMN1 0.7055124 0.0649667 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21175U20 -19 52 5

SMN1 1.4343330 0.0759369 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21175U20 -19 9 65

SMN1 0.5389321 0.3092735 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21175U20 -19 56 94

SMN1 1.1737463 0.1794157 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21175U20 -19 7 46

SMN1 1.1861414 0.0367290 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21175U20 -19 71 63

SMN1 1.8347753 0.1557617 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21175U20 -19 68 23

SMN1 0.8263034 0.0629982 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21176U20 -20 53 54 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 1.5057866 0.1706979 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21176U20 -20 89 84

SMN1 0.0624499 0.0490695 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21176U20 -20 2 71

SMN1 1.5414808 0.4611586 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21176U20 -20 55 69

SMN1 1.0899856 0.0437505 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21176U20 -20 92 68

SMN1 1.4153137 0.1465027 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21176U20 -20 5 26

SMN1 0.8655664 0.2094550 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21177U20 -21 53 26

SMN1 1.4666887 0.1162677 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21177U20 -21 87 64

SMN1 0.3882335 0.1396808 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21177U20 -21 14 69

SMN1 1.3662694 0.2394205 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21177U20 -21 47 57

SMN1 1.3545548 0.0131754 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21177U20 -21 41 63

SMN1 2.0269683 0.2782790 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21177U20 -21 82 2

SMN1 0.6399348 0.0116798 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21178U20 -22 51 91

SMN1 1.2425939 0.0284051 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21178U20 -22 23 9

SMN1 0.2298579 0.1281012 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21178U20 -22 22 82

SMN1 1.4997222 0.5687885 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21178U20 -22 55 39

SMN1 1.2347837 0.0171194 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21178U20 -22 64 32

SMN1 1.5096955 0.1757641 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21178U20 -22 91 56

SMN1 0.7480318 0.0837324 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21179U20 -23 45 79

SMN1 1.3391097 0.0708771 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21179U20 -23 3 43

SMN1 0.3841433 0.1472373 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21179U20 -23 84 5

SMN1 2.6201956 0.3421018 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21179U20 -23 11 26

SMN1 1.4736638 0.0537626 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21179U20 -23 66 05

SMN1 1.9208004 0.1273368 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21179U20 -23 18 42

SMN1 0.9074366 0.2420168 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21180U20 -24 01 1

SMN1 1.2837936 0.1586617 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21180U20 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 0.9631002 0.1178024 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21180U20 -24 08 22

SMN1 0.9947532 0.2684156 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21180U20 -24 99 48

SMN1 0.9654403 0.0326462 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21180U20 -24 48 95

SMN1 1.1405661 0.1016312 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21180U20 -24 71 1

SMN1 0.9088548 0.0760260 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21181U20 -25 08 35

SMN1 1.2261850 0.0444227 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21181U20 -25 41 05

SMN1 1.0550823 0.3267680 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21181U20 -25 01 36

SMN1 0.9691850 0.2264848 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21181U20 -25 38 66

SMN1 1.0097463 0.1221207 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21181U20 -25 6 37

SMN1 1.0813036 0.1018273 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21181U20 -25 39 03

SMN1 0.8764440 0.0704579 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21182U20 -26 72 09

SMN1 1.6324348 0.0615123 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21182U20 -26 88 57

SMN1 0.0715933 0.0715928 SMN1 in vitro Hep3B 50 qRTPCR SMN1:21182U20 -26 19 84

SMN1 1.9992025 0.4203876 SMN1 in vitro Hep3B 100 qRTPCR SMN1:21182U20 -26 16 69

SMN1 0.9741075 0.0668636 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21182U20 -26 84 61

SMN1 1.0302278 0.0961050 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21182U20 -26 91 98

SMN1 0.8343657 0.1081028 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21157U15 -27 03 71

SMN1 1.5899542 0.0931016 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21157U15 -27 19 53

SMN1 0.7471867 0.0078077 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21157U15 -27 14 01

SMN1 1.0490687 0.0926451 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21157U15 -27 44 93

SMN1 1.0583436 0.2089315 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21158U15 -28 94 76

SMN1 1.4023484 0.1019507 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21158U15 -28 14 71

SMN1 1.1502243 0.0800777 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21158U15 -28 16 07

SMN1 1.2198283 0.0317827 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21158U15 -28 96 62

SMN1 0.7122685 0.0775728 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21159U15 -29 87 38 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 1.1453055 0.0445753 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21159U15 -29 52 89

SMN1 0.9373938 0.0157007 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21159U15 -29 65 83

SMN1 1.2085219 0.0840218 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21159U15 -29 62 99

SMN1 0.8695041 0.1476827 SMN1 in vitro RPTEC 100 qRTPCR SMN1:21160U15 -30 09 79

SMN1 1.1669957 0.1289005 SMN1 in vitro RPTEC 50 qRTPCR SMN1:21160U15 -30 09 31

SMN1 1.0695334 0.0422583 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21160U15 -30 23 92

SMN1 1.0046189 0.0682455 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21160U15 -30 99 37

SMN1 1.2236852 0.1552583 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21161U15 -31 97 66

SMN1 0.9365695 0.0833678 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21161U15 -31 75 99

SMN1 1.0329784 0.0231205 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21162U15 -32 69 7

SMN1 1.0530458 0.0301583 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21162U15 -32 21 89

SMN1 1.0463614 0.0389718 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21163U15 -33 07 09

SMN1 1.2333022 0.0632553 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21163U15 -33 32 41

SMN1 1.0798767 0.0985940 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21164U15 -34 51 2

SMN1 1.2710261 0.0670194 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21164U15 -34 83 76

SMN1 0.8614640 0.0240959 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21165U15 -35 08 12

SMN1 0.8369663 0.0541596 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21165U15 -35 92 19

SMN1 1.2663632 0.0469636 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21166U15 -36 4 81

SMN1 1.3262571 0.0396746 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21166U15 -36 17 49

SMN1 1.2326900 0.0434762 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21167U15 -37 86 52

SMN1 1.1446329 0.0584333 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21167U15 -37 87 53

SMN1 0.8432418 0.0338080 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21168U15 -38 63 43

SMN1 0.9381803 0.0113762 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21168U15 -38 3 17

SMN1 0.6637462 0.0455270 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21169U15 -39 49 14

SMN1 0.8917645 0.0193953 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21169U15 -39 51 27 Oligo RQ RQ SE Gene Expt Type Cell [Oligo] Assay Coordinates_g Name Name Line/Tissue Type

SMN1 0.8881386 0.0814018 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21170U15 -40 53 04

SMN1 0.8716028 0.0653729 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21170U15 -40 99 36

SMN1 0.8824661 0.0310167 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21171U15 -41 48 49

SMN1 1.0936947 0.0259965 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21171U15 -41 65 02

SMN1 0.9568608 0.0435583 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21172U15 -42 36 82

SMN1 1.1517559 0.0676621 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21172U15 -42 99 07

SMN1 1.3419197 0.0808077 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21173U15 -43 82 6

SMN1 1.6929198 0.0846691 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21173U15 -43 15 98

SMN1 0 0 SMN1 NA NA 0 NA SMN1:21174U15 -44

SMN1 1.8072368 0.1141094 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21175U15 -45 97 8

SMN1 1.3777737 0.1085400 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21175U15 -45 03 58

SMN1 1.5456495 0.0640068 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21176U15 -46 38 14

SMN1 1.3542915 0.0384989 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21176U15 -46 04 44

SMN1 2.7115983 0.2600434 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21177U15 -47 61 46

SMN1 1.9866747 0.1194366 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21177U15 -47 86 75

SMN1 1.4823421 0.0630363 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21178U15 -48 95 43

SMN1 2.5973506 0.1454398 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21178U15 -48 28 01

SMN1 1.5344939 0.1106883 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21179U15 -49 05 65

SMN1 2.2233407 0.1487029 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21179U15 -49 84 92

SMN1 0.8974213 0.0342549 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21180U15 -50 96 31

SMN1 1.1323627 0.0785230 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21180U15 -50 81 03

SMN1 1.1579213 0.0442563 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21181U15 -51 68 19

SMN1 1.1776046 0.0380603 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21181U15 -51 65 53

SMN1 0.9735483 0.0514615 SMN1 in vitro Hep3B 30 qRTPCR SMN1:21182U15 -52 53 83

SMN1 1.0683556 0.0608511 SMN1 in vitro Hep3B 10 qRTPCR SMN1:21182U15 -52 42 46 Table 3: Oligonucleotide Modifications

Symbol Feature Description

bio 5' biotin

dAs DNA w/3' thiophosphate dCs DNA w/3' thiophosphate dGs DNA w/3' thiophosphate dTs DNA w/3' thiophosphate dG DNA

enaAs EN A w/3' thiophosphate enaCs EN A w/3' thiophosphate enaGs EN A w/3' thiophosphate enaTs EN A w/3' thiophosphate fluAs 2'-fhioro w/3' thiophosphate fluCs 2'-fluoro w/3' thiophosphate fluGs 2'-fluoiO w/3' thiophosphate fluUs 2'-fluoro w/3' thiophosphate

InaAs LNA w/3' thiophosphate

InaCs LNA w/3' thiophosphate

InaGs LNA w/3' thiophosphate

InaTs LNA w/3' thiophosphate omeAs 2'-OMe w/3' thiophosphate omeCs 2'-OMe w/3' thiophosphate omeGs 2'-OMe w/3' thiophosphate omeTs 2'-OMe w/3' thiophosphate

InaAs-Sup LNA w/3' thiophosphate at 3' terminus

InaCs-Sup LNA w/3' thiophosphate at 3' terminus

InaGs-Sup LNA w/3' thiophosphate at 3' terminus

InaTs-Sup LNA w/3' thiophosphate at 3' terminus

InaA-Sup LNA w/3' OH at 3' terminus

InaC-Sup LNA w/3' OH at 3' terminus

InaG-Sup LNA w/3' OH at 3' terminus

InaT-Sup LNA w/3' OH at 3' terminus omeA-Sup 2'-OMe w/3' OH at 3' terminus omeC-Sup 2'-OMe w/3' OH at 3' terminus omeG-Sup 2'-OMe w/3' OH at 3' terminus omeU-Sup 2'-OMe w/3' OH at 3' terminus moeAs 2'-O-MOE w/3' thiophosphate moeCs 2 -O-MOE w/3' thiophosphate moeGs 2'-O-MOE w/3' thiophosphate moeTs 2 -O-MOE w/3' thiophosphate Symbol Feature Description

dAs-Sup DNA w/3' thiophosphate at 3' terminus

dCs-Sup DNA w/3' thiophosphate at 3' terminus

dGs-Sup DNA w/3' thiophosphate at 3' terminus

dTs-Sup DNA w/3' thiophosphate at 3' terminus

dA-Sup DNA w/3' OH at 3' terminus

dC-Sup DNA w/3' OH at 3' terminus

dG-Sup DNA w/3' OH at 3' terminus

dT-Sup DNA w/3' OH at 3' terminus

BRIEF DESCRIPTION OF SEQUENCE LISTING

SeqID Chrom Gene Chrom Start Chrom End Strand Name

1 chr5 SMN1 70208768 70260838 + human SMN1

2 chr5 SMN2 69333350 69385422 + human SMN2

3 chr9 SMNP 20319406 20344375 + human SMNP

4 chr5 SMN1 70208768 70260838 - human SMNl_revComp

5 chr5 SMN2 69333350 69385422 - human SMN2_revComp

6 chr9 SMNP 20319406 20344375 - human SMNP_revComp

7 chrl3 Smnl 100881160 100919653 + mouse Smnl

8 chrl3 Smnl 100881160 100919653 - mouse Smnl_revComp

9 chr5 SMN1 70240095 70240127 + S48-193240

9 chr5 SMN2 69364672 69364704 + S48-193240

10 chr5 SMN1 70214393 70214822 + S48-441814

10 chr5 SMN2 69338976 69339405 + S48-441814

11 chr5 SMN1 70214064 70214108 + S48-441815

11 chr5 SMN2 69338647 69338691 + S48-441815

12 chr5 SMN1 70214276 70214317 + S48-473289

12 chr5 SMN2 69338859 69338900 + S48-473289

13 chr5 SMN1 70214445 70214472 + S48-473290

13 chr5 SMN2 69339028 69339055 + S48-473290

14 chr5 SMN1 70238095 70242127 + S48-193240 +2K

15 chr5 SMN2 69362672 69366704 + S48-193240 +2K

16 chr5 SMN1 70212393 70216822 + S48-441814 +2K

17 chr5 SMN2 69336976 69341405 + S48-441814 +2K

18 chr5 SMN1 70212064 70216108 + S48-441815 +2K

19 chr5 SMN2 69336647 69340691 + S48-441815 +2K

20 chr5 SMN1 70212276 70216317 + S48-473289 +2K

21 chr5 SMN2 69336859 69340900 + S48-473289 +2K

22 chr5 SMN1 70212445 70216472 + S48-473290 +2K 23 chr5 SMN2 69337028 69341055 + S48-473290 +2K

24 chr5 SMN1 70240510 70240551 - S48-193241

24 chr5 SMN2 69365087 69365128 - S48-193241

25 chr5 SMN1 70241924 70241968 - S48-193242

25 chr5 SMN2 69366499 69366543 - S48-193242

26 chr5 SMN1 70238510 70242551 - S48-193241 +2K

27 chr5 SMN2 69363087 69367128 - S48-193241 +2K

28 chr5 SMN1 70239924 70243968 - S48-193242 +2K

29 chr5 SMN2 69364499 69368543 - S48-193242 +2K

13100 chr5 SMN1 70247831 70247845 + Splice control sequence

13100 chr5 SMN2 69372411 69372425 + Splice control sequence

13101 chr5 SMN2 69372402 69372845 + Intron 7

Single Strand Oligonucleotides (Antisense Strand of Target Gene)

SeqID range: 30 to 8329, 13088-13094 Example SeqIDs w/o G Runs: 30-142, 156-560, 575-780, 794-912, 926-1013, 1027-1078, 1092-1286, 1300-1335, 1349- 1385, 1399-1453, 1460-1527, 1548-1555, 1571-1653, 1675-1691, 1706-1802, 1816-1883, 1897-2009, 2023-2141, 2165-2289, 2303-2320, 2334-2447, 2461-2494, 2508-2526, 2540- 2545, 2571-2635, 2651-2670, 2689-2763, 2772-2814, 2828-2854, 2868-3030, 3044-3256, 3270-3360, 3374-3400, 3414-3722, 3737, 3759-3783, 3797-3970, 3986-4059, 4073-4153, 4175-4240, 4255-4415, 4438-4441, 4456-4472, 4484-4505, 4513-4516, 4531-4546, 4560- 4650, 4664-4751, 4766-4918, 4932-5035, 5049-5064, 5091-5189, 5203-5448, 5459-5503, 5508-5520, 5535-5654, 5668-5863, 5877-6016, 6025-6029, 6054-6063, 6078-6215, 6229- 6701, 6715-6729, 6744-6869, 6883-6945, 6959-6968, 6982-7085, 7099-7173, 7195-7247, 7255-7268, 7273-7309, 7320-7335, 7349-7442, 7456-7465, 7479-7727, 7740-7951, 7977- 8208, 8223-8255, 8257-8296, 8304-8312, 8319-8329, 13093-13094

Example SeqIDs w/o miR Seeds:

30-32, 34-39, 45-62, 64-72, 77-142, 145-151, 153, 157-184, 186-202, 205-246, 249, 251-260, 263, 266-320, 322, 326-328, 331, 333-341, 343-344, 346-394, 396-445, 447-541, 543-562, 564-596, 599-605, 607-646, 648-673, 677-701, 703-735, 737-772, 774-781, 785, 787-793, 795-809, 812, 814-815, 819-820, 822-827, 833-834, 836, 838-850, 852-876, 879, 883-886, 889-890, 892, 894, 897, 899, 901-906, 909, 911, 919, 921-935, 940, 942-1012, 1016-1063, 1065-1067, 1069-1095, 1097-1147, 1149-1166, 1168-1190, 1193-1214, 1217, 1227-1237, 1240, 1244, 1246-1251, 1258-1281, 1283-1312, 1314-1333, 1335, 1337-1356, 1358-1364, 1367, 1369-1381, 1384, 1388-1389, 1392-1404, 1406-1417, 1421-1441, 1443, 1445, 1447- 1460, 1462-1492, 1494-1500, 1502, 1506-1512, 1514-1531, 1533, 1535-1539, 1543-1558, 1561-1562, 1564-1601, 1603-1614, 1616-1633, 1635-1646, 1648-1656, 1658, 1661-1675, 1678-1716, 1718-1740, 1742-1750, 1752-1785, 1788-1795, 1798, 1804-1871, 1873-1884, 1892-1973, 1976-1992, 1998-2032, 2034-2053, 2055-2077, 2079-2116, 2118-2135, 2138, 2142, 2149, 2151-2153, 2155-2162, 2165-2174, 2178, 2181-2254, 2256-2268, 2271-2293, 2296-2298, 2301-2312, 2314-2323, 2325-2427, 2429-2438, 2441, 2445, 2450-2476, 2478- 2489, 2492-2494, 2497-2513, 2515, 2521-2526, 2529-2545, 2547-2571, 2573-2610, 2613- 2620, 2622-2639, 2641, 2644, 2651-2743, 2745, 2747-2755, 2760-2775, 2777-2825, 2828- 2841, 2844-2861, 2864-2888, 2894-2954, 2956-2988, 2991-3006, 3008-3043, 3046, 3048- 3239, 3241-3253, 3256-3268, 3270-3273, 3276-3320, 3322-3355, 3357-3404, 3406-3428, 3430-3488, 3490-3491, 3493-3522, 3524-3552, 3554-3569, 3571-3650, 3653-3670, 3672- 3688, 3690-3717, 3719-3724, 3727-3736, 3743, 3746, 3749, 3751-3821, 3823-3842, 3844, 3846, 3848, 3851, 3855, 3858, 3861-3863, 3866-3881, 3883, 3887-3907, 3909-3917, 3919- 3924, 3926-3942, 3944-3952, 3956-3970, 3976-3988, 3991-3998, 4001-4008, 4010-4022, 4026, 4028-4039, 4041-4048, 4051-4059, 4063-4070, 4072-4073, 4075, 4078, 4080-4104, 4106-4124, 4126, 4129-4140, 4143-4153, 4156-4162, 4166-4171, 4174-4196, 4201-4241, 4244-4252, 4254-4285, 4289-4318, 4320-4324, 4327, 4330-4331, 4333-4335, 4337-4351, 4353-4414, 4417, 4419-4425, 4427-4433, 4435, 4438-4446, 4449-4450, 4452-4462, 4470- 4472, 4474-4510, 4512-4517, 4520-4546, 4548, 4551-4590, 4592-4619, 4623-4696, 4699, 4701, 4703-4752, 4755-4877, 4879-4949, 4951-5007, 5010, 5013-5046, 5049-5059, 5061- 5063, 5066, 5069-5078, 5080-5119, 5121-5131, 5134-5168, 5171-5189, 5191-5228, 5230- 5341, 5343-5405, 5407-5426, 5429-5437, 5439-5476, 5478-5491, 5494-5516, 5518-5556, 5558-5572, 5574-5647, 5649, 5652-5653, 5657-5729, 5731-5742, 5744-5769, 5771-5780, 5782-5866, 5868, 5870-5876, 5879-5881, 5884-5899, 5902-5951, 5954-5993, 6000-6006, 6009-6016, 6019-6033, 6035-6065, 6067-6073, 6080-6165, 6168-6249, 6252-6257, 6261- 6282, 6284-6289, 6291-6301, 6305-6367, 6369-6378, 6380-6398, 6401-6412, 6415, 6417- 6447, 6449-6456, 6458-6500, 6502-6563, 6565-6589, 6591-6612, 6616-6660, 6663-6702, 6704-6748, 6753, 6758-6763, 6765, 6768-6807, 6810, 6812-6872, 6874-6877, 6879-6913, 6915-6916, 6919, 6922, 6924-6926, 6928, 6930-6936, 6940-6959, 6962, 6964-6990, 6992, 6996, 6998-6999, 7004-7038, 7042-7085, 7087, 7089, 7092-7134, 7136-7140, 7142-7143, 7146-7150, 7152-7157, 7159-7171, 7175, 7177-7178, 7180-7196, 7198-7220, 7223, 7231- 7237, 7239, 7242-7246, 7250-7273, 7275-7308, 7310-7312, 7314-7317, 7319-7330, 7332- 7400, 7402-7437, 7439, 7441-7466, 7470-7491, 7493, 7495, 7497, 7499, 7502-7614, 7622- 7628, 7631-7646, 7649-7651, 7655-7657, 7661-7672, 7676, 7679-7721, 7723-7800, 7802- 7803, 7805-7906, 7908, 7910-7939, 7943-7953, 7956-7964, 7966-7981, 7983, 7985-7999, 8002, 8004-8034, 8036-8046, 8048-8080, 8084-8094, 8096-8112, 8114-8115, 8117-8139, 8141-8143, 8146-8148, 8150-8187, 8190-8216, 8218-8229, 8232-8238, 8240-8250, 8253- 8255, 8257-8275, 8278-8296, 8299-8304, 8306-8329, 13093-13094

Single Strand Oligonucleotides (Sense Strand of Target Gene)

SeqID range: 1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2546, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-13061, 13062-13087, 13108-13116

Example SeqIDs w/o G Runs:

1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2490, 2542-2545, 2656-2657, 2833-2835, 3439-3440, 3916-3918, 4469-4472, 4821, 5429, 5537, 6061, 7327, 8330-8495, 8520-8560, 8574-8837, 8857-8882, 8907-8964, 8978-9298, 9312-9382, 9394-9640, 9656- 9753, 9767-9974, 9988-10261, 10275-10276, 10290-10301, 10315-10434, 10448-10613, 10623-10641, 10644-10676, 10678-10704, 10714-10802, 10822-11161, 11175-11192, 11207-11386, 11400-11730, 11744-11745, 11759-11852, 11857-11900, 11914-11984, 11999-12011, 12026-12153, 12163-12175, 12178-12195, 12198-12212, 12216-12536, 12547-12564, 12575-12664, 12674-12758, 12772-12797, 12800-12840, 12854-13061, 13062-13069

Example SeqIDs w/o miR Seeds:

1158-1159, 1171, 1482-1483, 1485-1486, 2465-2471, 2488-2489, 2542-2545, 2656-2657, 2833-2835, 3439-3440, 3916-3917, 4470-4472, 4821, 5429, 5537, 6061, 7327, 8330-8334, 8336-8345, 8347, 8351-8373, 8375-8390, 8392-8399, 8401-8413, 8415-8455, 8457-8493, 8495, 8497-8502, 8510-8517, 8520, 8525, 8527-8634, 8637-8653, 8655-8671, 8673-8718, 8721-8822, 8824-8825, 8827-8842, 8849-8879, 8881-8892, 8894-8902, 8905-8906, 8914- 8927, 8929, 8931, 8935, 8937-8975, 8980-8992, 8994, 8996-8997, 8999-9001, 9003, 9005- 9086, 9089-9124, 9126-9286, 9288-9307, 9310-9359, 9362-9420, 9425-9427, 9429-9432, 9434, 9436-9437, 9439-9461, 9464-9483, 9486, 9488-9498, 9500-9511, 9513, 9515-9650, 9653-9667, 9669, 9671-9723, 9725-9869, 9871-9872, 9874-9879, 9881-9889, 9891-9973, 9975-10077, 10080-10097, 10099, 10101-10127, 10129-10166, 10168-10170, 10172-10184, 10186-10230, 10232-10237, 10239-10260, 10262-10272, 10274-10342, 10344-10400, 10402-10423, 10426-10441, 10445-10556, 10560, 10562-10580, 10582-10606, 10609- 10647, 10650, 10652-10704, 10706, 10710-10713, 10716-10731, 10733-10824, 10826- 10842, 10844-10903, 10906-10907, 10909-11101, 11104, 11106-11134, 11137-11138, 11145-11161, 11164-11173, 11175-11181, 11184, 11186-11203, 11207-11212, 11214- 11239, 11243-11259, 11261-11347, 11351-11397, 11399-11740, 11742-11747, 11749- 11790, 11792-11817, 11821-11852, 11854-11904, 11908-11944, 11946-11959, 11961, 11964-11984, 11986-12007, 12009-12022, 12024-12092, 12095-12119, 12121-12133, 12135-12144, 12146-12157, 12159-12225, 12227-12231, 12233-12300, 12302-12329, 12332-12333, 12335-12382, 12385-12411, 12414-12416, 12418-12444, 12446-12455, 12457-12461, 12465, 12468-12474, 12476-12499, 12501-12536, 12538-12544, 12547- 12553, 12559-12610, 12612-12626, 12628-12631, 12633-12637, 12640-12645, 12648- 12657, 12659-12671, 12673-12679, 12683-12710, 12712, 12714-12747, 12750, 12752- 12766, 12769, 12771-12806, 12808-12826, 12828-12829, 12831, 12833-12846, 12848- 12849, 12854-12931, 12933-12946, 12948-13061, 13064, 13066-13068, 13071-13072, 13075, 13077-13081, 13083, 13085, 13087

Example 2: Selective upregulation ofexon 7 containing SMN2 transcripts using oligonucleotides targeting PRC2-interacting regions that upregulate SMN2 and splice- switching oligonucleotides.

Oligo design:

Oligonucleotides targeting PRC2-interacting regions (IncRNA peaks) in the SMN1/2 gene loci were designed. These oligos were synthesized with various DNA base modifications, modification placements, inter-nucleoside bonds and inter-oligo linkers (oligos 1-52 and 59-101) as outlined in Table 4.

Splice switching oligos (SSO) were designed based on sequences of SMN2. Various modifications of such SSOs in length and chemistry were prepared (oligos 53-58).

Universal negative control oligos (oligo 232 and 293) were also designed using on bioinformatic analysis.

Methods:

Cell Culture:

Six fibroblast cell lines and one lymphoblast cell line were obtained from the Coriell Institute (FIG. 2). The cells were either transfected with the oligos using Lipofectamine 2000

(Fibroblasts) or by electroporation or unassisted delivery (lymphoblast) to ascertain effects of the oligonucleotides on SMN1/2 mRNA and protein expression. All experiments were carried out as biological triplicates.

mRNA and protein expression:

mRNA expression -

1. On day 1, patient fibroblasts with reduced SMN expression were seeded into each well of 96- well plates at a density of 5,000 cells per lOOuL.

2. On day 2, transfections were performed using Lipofectamie2000 per manufacturer's instructions with oligos at either ΙΟηΜ or 30nM.

3. 48 hours post-transfection, Ambion Cells-to-CT kit was used to directly obtain qRT-PCR results from the cells per manufacturer's instructions.

4. Quantitative PCR evaluation was completed using Taqman FAST qPCR on StepOne Plus, and change in mRNA expression was quantified using the delta delta Ct method by normalizing SMN expression to a housekeeper gene (B2M).

Protein expression (ELISA) -

ELISA to determine SMN protein was carried out per manufacturer's instructions (SMN ELISA kit #ADI-900-209, Enzo Life Sciences). Briefly, SMN fibroblasts were cultures at 40,000 cells/well of a 24- well tissue culture coated plate on day 1. Cells were transfected with the oligos using Lipofectamine2000 on day 2 and cell lysates prepared at 24 and 48 hours post-treatment. ELISA was carried out per manufacturer's instructions.

Subsequently fold induction of SMN protein was determined by normalizing SMN protein levels induced by oligonucleotides to the SMN protein levels induced by Lipofectamine treatment alone.

Splice switching assay (Ddel assay) -

SMN2-derived transcripts contain a unique Ddel restriction element introduced because of a nucleotide polymorphism not present in SMN1 and are differentiated from SMN 1 -derived transcripts because of the faster migration of the SMN2 products. Briefly, patient fibroblasts with reduced SMN expression were treated with oligonucleotides targeting PRC2-interacting regions with or without SSO at 30nM each as described before. RT-PCR was carried out with an SMN exon 5 forward primer and an exon 8 reverse primer to generate cDNAs that were then digested with Ddel. The SMN1 transcript, if present, migrates at a slower rate than the Ddel-digested SMN2 transcript and is seen as the first band from the top of the gel. The second band from the top indicates full length SMN2 (accurately spliced form) and the third band indicates the incorrectly spliced SMN2delta7. (FIG. 5)

Results

Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are both neurodegenerative diseases in which motor neurons degenerate and die, leading to muscle weakness and ultimately paralysis. In both cases, motor neurons exhibit defects in spliceosome integrity as indicated by disruption of Gems (Gemini of coiled bodies).

In some embodiments, the SMN1 gene is mutated in such a way that it is unable to correctly code the SMN protein - due to either a deletion encompassing at least a portion of exon 7 or to other mutations. SMA patients, however, generally retain at least one copy of the SMN2 gene (with many having 2 or more copies) that still expresses small amounts of SMN protein. The SMN2 gene has a C to T mutation (compared with SMN1) in exon 7 that alters splicing of its precursor mRNA such that exon 7 is spliced out at a high frequency. Consequently, only about 10% of the normal levels of full length SMN protein are produced from SMN2. (See FIG. 1)

Six SMA fibroblast cell lines and one lymphoblast cell line were obtained from the Coriell Institute (FIG. 2). Cells were transfected with oligonucleotides (oligos 1-52 and 59- 101) directed against a PRC2-associated region of SMN2 and RT-PCR assays were conducted to evaluate effects on SMN mRNA expression. (See FIGS. 3 and 4 for results in cell lines 3814 and 3813). It is expected that oligos directed againsted a PRC2-associated region of SMN may further enhance SMN protein expression when wild- type SMN1 is present, e.g., in cells from subjects with ALS. In separate experiments, cells were transfected with oligonucleotides (oligos 53-58) directed at a splice control sequence in intron 7 of SMN2 and RT-PCR assays were conducted to evaluate effects on SMN mRNA expression (See FIGS. 3 and 4 for results in cell lines 3814 and 3813). Splice switching oligonucleotides (oligos 53-58) were found to increase expression of full length SMN2 based on a gel separation analysis of PCR products obtained following a Ddel restriction digest; whereas certain oligonucleotides directed against a PRC2-associated region of SMN2 did not. (FIG. 5)

SMN ELISA (Enzo) assays were conducted and revealed that certain oligonucleotides directed against a PRC2-associated region of SMN2 alone did not significantly increase full length SMN protein 24h post-transfection in certain SMA patient fibroblasts. (FIG. 6) However, the same SMN ELISA assays showed that oligonucleotides directed against a PRC2-associated region of SMN2 in combination with a splice switching oligonucleotide (oligo 53 or 54) significantly increase full length SMN protein 24h post-transfection in SMA patient fibroblasts above that observed with splice switching oligonucleotides alone. (FIGS. 7 and 8). RT-PCR assays were conducted and showed that oligonucleotides directed against a PRC2-associated region of SMN2 in combination with a splice switching oligonucleotide (oligo 53 or 54) significantly increased SMN2 protein 24h post-transfection in SMA patient fibroblasts. (FIG 9.) These experiments were conducted modified oligonucleotides with either alternating LNA and 2'OMe nucleotides or alternating DNA and LNA nucleotides.

In summary, the results of Example 2 show that certain oligos targeting PRC2 associated regions of SMN2 induce SMN RNA expression (e.g.., of the SMNA7 transcript) SMA patient-derived fibroblasts. The results also show that, in some embodiments, splice- switching oligos may not induce SMN RNA expression, but rather shift SMN RNA splicing to the full-length transcript. Finally, the results show that combination of splice switching oligos with PRC2-associated region targeting oligos substantially increases full length SMN protein in cells from SMA patients.

Table 4: Oligonucleotide sequences made for testing human cells obtained from subjects with Spinal Muscular Atrophy (See Table 3 for structural features of formatted sequence).

Oligo Base Formatted Sequence SeqID

Name Sequence SM N l- ATTCTCTTGA omeAs;omeUs;omeUs;omeCs;omeUs;omeCs;omeUs;omeUs;omeGs;om 13062 1 m03 TGATGCTGAT eAs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs

;omeU-Sup

SM N 1- TTCTCTTGAT omeUs;omeUs;omeCs;omeUs;omeCs;omeUs;omeUs;omeGs;omeAs;om 13063 2 m03 GATGCTGAT eUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs

G ;omeG-Sup

SM N 1- TCTCTTGATG omeUs;omeCs;omeUs;omeCs;omeUs;omeUs;omeGs;omeAs;omeUs;om 13064 3 m03 ATGCTGATGC eGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs

;omeC-Sup

SM N 1- CTCTTGATGA omeCs;omeUs;omeCs;omeUs;omeUs;omeGs;omeAs;omeUs;omeGs;om 13065 4 m03 TGCTGATGCT eAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs

;omeU-Sup

SM N 1- TCTTGATGAT omeUs;omeCs;omeUs;omeUs;omeGs;omeAs;omeUs;omeGs;omeAs;om 13066 5 m03 GCTGATGCTT eUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs

;omeU-Sup

SM N 1- CTTGATGATG omeCs;omeUs;omeUs;omeGs;omeAs;omeUs;omeGs;omeAs;omeUs;om 13067 6 m03 CTGATGCTTT eGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs

;omeU-Sup

SM N 1- TTGATGATGC omeUs;omeUs;omeGs;omeAs;omeUs;omeGs;omeAs;omeUs;omeGs;om 13068 7 m03 TGATGCTTTG eCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs

;omeG-Sup

SM N 1- TGATGATGCT omeUs;omeGs;omeAs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;om 13069 8 m03 GATGCTTTGG eUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs

;omeG-Sup

SM N 1- GATGATGCT omeGs;omeAs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;om 13070 9 m03 GATGCTTTGG eGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs

G ;omeG-Sup

SM N 1- ATGATGCTGA omeAs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;om 13071

10 m03 TGCTTTGGGA eAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs

;omeA-Sup

SM N 1- TGATGCTGAT omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;om 13072

11 m03 GCTTTGGGA eUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs

A ;omeA-Sup

SM N 1- GATGCTGAT omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;om 13073

12 m03 GCTTTGGGA eGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs

AG ;omeG-Sup

SM N 1- ATGCTGATGC omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;om 13074

13 m03 TTTGGGAAGT eCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs

;omeU-Sup

SM N 1- TGCTGATGCT omeUs;omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;om 13075

14 m03 TTGGGAAGT eUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs

A ;omeA-Sup

SM N 1- GCTGATGCTT omeGs;omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;om 13076

15 m03 TGGGAAGTA eUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs

T ;omeU-Sup

SM N 1- CTGATGCTTT omeCs;omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;om 13077

16 m03 GGGAAGTAT eUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs;omeUs

G ;omeG-Sup

SM N 1- TGATGCTTTG omeUs;omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;om 13078

17 m03 GGAAGTATG eGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs;omeUs;omeGs

T ;omeU-Sup SM N l- GATGCTTTGG omeGs;omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;om 13079

18 m03 GAAGTATGTT eGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs;omeUs;omeGs;omeUs

;omeU-Sup

SM N 1- ATGCTTTGGG omeAs;omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;om 13080

19 m03 AAGTATGTTA eGs;omeAs;omeAs;omeGs;omeUs;omeAs;omeUs;omeGs;omeUs;omeUs

;omeA-Sup

SM N 1- TGCTTTGGGA omeUs;omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;om 13081 0 m03 AGTATGTTAA eAs;omeAs;omeGs;omeUs;omeAs;omeUs;omeGs;omeUs;omeUs;omeAs

;omeA-Sup

SM N 1- GCTTTGGGA omeGs;omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;om 13082 1 m03 AGTATGTTAA eAs;omeGs;omeUs;omeAs;omeUs;omeGs;omeUs;omeUs;omeAs;omeAs

T ;omeU-Sup

SM N 1- CTTTGGGAA omeCs;omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;om 13083 2 m03 GTATGTTAAT eGs;omeUs;omeAs;omeUs;omeGs;omeUs;omeUs;omeAs;omeAs;omeUs

T ;omeU-Sup

SM N 1- TTTGGGAAGT omeUs;omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;om 13084 3 m03 ATGTTAATTT eUs;omeAs;omeUs;omeGs;omeUs;omeUs;omeAs;omeAs;omeUs;omeUs

;omeU-Sup

SM N 1- TTGGGAAGT omeUs;omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;om 13085 4 m03 ATGTTAATTT eAs;omeUs;omeGs;omeUs;omeUs;omeAs;omeAs;omeUs;omeUs;omeUs

C ;omeC-Sup

SM N 1- TGGGAAGTA omeUs;omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs;om 13086 5 m03 TGTTAATTTC eUs;omeGs;omeUs;omeUs;omeAs;omeAs;omeUs;omeUs;omeUs;omeCs

A ;omeA-Sup

SM N 1- GGGAAGTAT omeGs;omeGs;omeGs;omeAs;omeAs;omeGs;omeUs;omeAs;omeUs;om 13087 6 m03 GTTAATTTCA eGs;omeUs;omeUs;omeAs;omeAs;omeUs;omeUs;omeUs;omeCs;omeAs

T ;omeU-Sup

SM N 1- ATTCTCTTGA lnaAs;omeUs;lnaTs;omeCs;lnaTs;omeCs;lnaTs;omeUs;lnaGs;omeAs; lnaT 11374 7 mOl TGATG s;omeGs;lnaAs;omeUs;lnaG-Sup

SM N 1- TTCTCTTGAT lnaTs;omeUs;lnaCs;omeUs;lnaCs;omeUs;lnaTs;omeGs;lnaAs;omeUs; lna 11375 8 mOl GATGC Gs;omeAs;lnaTs;omeGs;lnaC-Sup

SM N 1- TCTCTTGATG lnaTs;omeCs;lnaTs;omeCs;lnaTs;omeUs;lnaGs;omeAs;lnaTs;omeGs; lnaA 11376 9 mOl ATGCT s;omeUs;lnaGs;omeCs;lnaT-Sup

SM N 1- CTCTTGATGA lnaCs;omeUs;lnaCs;omeUs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeAs; lnaT 11377 0 mOl TGCTG s;omeGs;lnaCs;omeUs;lnaG-Sup

SM N 1- TCTTGATGAT lnaTs;omeCs;lnaTs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaAs;omeUs; lnaG 11378 1 mOl GCTGA s;omeCs;lnaTs;omeGs;lnaA-Sup

SM N 1- CTTGATGATG lnaCs;omeUs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeAs;lnaTs;omeGs; lnaC 11379 2 mOl CTGAT s;omeUs;lnaGs;omeAs;lnaT-Sup

SM N 1- TTGATGATGC lnaTs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs; lnaT 11380 3 mOl TGATG s;omeGs;lnaAs;omeUs;lnaG-Sup

SM N 1- TGATG ATGCT lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs; lna 11381 4 mOl GATGC Gs;omeAs;lnaTs;omeGs;lnaC-Sup

SM N 1- G ATG ATGCT lnaGs;omeAs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeGs; lnaA 11382 5 mOl GATGCT s;omeUs;lnaGs;omeCs;lnaT-Sup

SM N 1- ATGATGCTGA lnaAs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeAs; lna 11383 6 m01 TGCTT Ts;omeGs;lnaCs;omeUs;lnaT-Sup

SM N 1- TGATG CTGAT lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeGs;lnaAs;omeUs; lna 11384 7 mOl GCTTT Gs;omeCs;lnaTs;omeUs;lnaT-Sup

SM N 1- GATGCTGAT lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeAs;lnaTs;omeGs; lnaC 11385 8 mOl GCTTTG s;omeUs;lnaTs;omeUs;lnaG-Sup SM N l- ATGCTGATGC lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs; lnaT 11386 39 mOl TTTGG s;omeUs;lnaTs;omeGs;lnaG-Sup

SM N 1- TGCTGATGCT lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs; lnaT 11387 0 mOl TTGGG s;omeUs;lnaGs;omeGs;lnaG-Sup

SM N 1- GCTGATGCTT lnaGs;omeCs;lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeUs; lnaT 11388 1 mOl TGGGA s;omeGs;lnaGs;omeGs;lnaA-Sup

SM N 1- CTGATGCTTT lnaCs;omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaTs;omeUs; lna 11389 2 mOl GGGAA Gs;omeGs;lnaGs;omeAs;lnaA-Sup

SM N 1- TGATGCTTTG lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeUs;lnaTs;omeGs; lnaG 11390 3 mOl GGAAG s;omeGs;lnaAs;omeAs;lnaG-Sup

SM N 1- GATGCTTTGG lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaTs;omeUs;lnaGs;omeGs; lna 11391 4 mOl GAAGT Gs;omeAs;lnaAs;omeGs;lnaT-Sup

SM N 1- ATGCTTTGGG lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeUs;lnaTs;omeGs;lnaGs;omeGs; lna 11392 5 mOl AAGTA As;omeAs;lnaGs;omeUs;lnaA-Sup

SM N 1- TGCTTTGGGA lnaTs;omeGs;lnaCs;omeUs;lnaTs;omeUs;lnaGs;omeGs;lnaGs;omeAs; lna 11393 6 mOl AGTAT As;omeGs;lnaTs;omeAs;lnaT-Sup

SM N 1- GCTTTGGGA dGs;lnaCs;dTs;lnaTs;dTs;lnaGs;dGs;lnaGs;dAs;lnaAs;dGs;lnaTs; dAs;lnaTs; 11394 7 m02 AGTATG dG-Sup

SM N 1- GCTTTGGGA lnaGs;omeCs;lnaTs;omeUs;lnaTs;omeGs;lnaGs;omeGs;lnaAs;omeAs; lna 11394 7 mOl AGTATG Gs;omeUs;lnaAs;omeUs;lnaG-Sup

SM N 1- CTTTGGGAA dCs;lnaTs;dTs;lnaTs;dGs;lnaGs;dGs;lnaAs;dAs;lnaGs;dTs;lnaAs; dTs;lnaGs; 11395 8 m05 GTATGT dT-Sup

SM N 1- CTTTGGGAA lnaCs;omeUs;lnaTs;omeUs;lnaGs;omeGs;lnaGs;omeAs;lnaAs;omeGs; lna 11395 8 mOl GTATGT Ts;omeAs;lnaTs;omeGs;lnaT-Sup

SM N 1- TTTGGGAAGT lnaTs;omeUs;lnaTs;omeGs;lnaGs;omeGs;lnaAs;omeAs;lnaGs;omeUs; lna 11396 9 mOl ATGTT As;omeUs;lnaGs;omeUs;lnaT-Sup

SM N 1- TTGGGAAGT lnaTs;omeUs;lnaGs;omeGs;lnaGs;omeAs;lnaAs;omeGs;lnaTs;omeAs; lnaT 11397 0 mOl ATGTTA s;omeGs;lnaTs;omeUs;lnaA-Sup

SM N 1- TGGGAAGTA lnaTs;omeGs;lnaGs;omeGs;lnaAs;omeAs;lnaGs;omeUs;lnaAs;omeUs; lna 11398 1 mOl TGTTAA Gs;omeUs;lnaTs;omeAs;lnaA-Sup

SM N 1- GGGAAGTAT lnaGs;omeGs;lnaGs;omeAs;lnaAs;omeGs;lnaTs;omeAs;lnaTs;omeGs; lnaT 11399 2 mOl GTTAAT s;omeUs;lnaAs;omeAs;lnaT-Sup

SM N 1- TCACTTTCAT dTs;lnaCs;dAs;lnaCs;dTs;lnaTs;dTs;lnaCs;dAs;lnaTs;dAs;lnaAs; dTs;lnaGs; 13088 3 m02 AATGCTGG dCs;lnaTs;dGs;lnaG-Sup

SM N 1- TCACTTTCAT lnaTs;dCs;lnaAs;dCs;lnaTs;dTs;lnaTs;dCs;lnaAs;dTs;lnaAs;dAs; lnaTs;dGs;l 13088 3 ml2 AATGCTGG naCs;dTs;lnaGs;dG-Sup

SM N 1- TCACTTTCAT lnaTs;omeCs;lnaAs;omeCs;lnaTs;omeUs;lnaTs;omeCs;lnaAs;omeUs; lnaA 13088 4 mOl AATGCTGG s;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeG-Sup

SM N 1- TCACTTTCAT omeUs;omeCs;omeAs;omeCs;omeUs;omeUs;omeUs;omeCs;omeAs;ome 13088 3 m03 AATGCTGG Us;omeAs;omeAs;omeUs;omeGs;omeCs;omeUs;omeGs;omeG-Sup

SM N 1- TCACTTTCAT lnaTs;omeCs;lnaAs;omeCs;lnaTs;omeUs;lnaTs;omeCs;lnaAs;omeUs; lnaA 13089 5 mOl AATGC s;omeAs;lnaTs;omeGs;lnaC-Sup

SM N 1- CACTTTCATA lnaCs;omeAs;lnaCs;omeUs;lnaTs;omeUs;lnaCs;omeAs;lnaTs;omeAs; lnaA 13090 6 m01 ATGCT s;omeUs;lnaGs;omeCs;lnaT-Sup

SM N 1- ACTTTCATAA dAs;lnaCs;dTs;lnaTs;dTs;lnaCs;dAs;lnaTs;dAs;lnaAs;dTs;lnaGs; dCs;lnaTs; 13091 7 m02 TGCTG dG-Sup

SM N 1- ACTTTCATAA lnaAs;omeCs;lnaTs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeAs; lnaT 13091 7 mOl TGCTG s;omeGs;lnaCs;omeUs;lnaG-Sup

SM N 1- CTTTCATAAT lnaCs;omeUs;lnaTs;omeUs;lnaCs;omeAs;lnaTs;omeAs;lnaAs;omeUs; lnaG 13092 8 mOl GCTGG s;omeCs;lnaTs;omeGs;lnaG-Sup

SM N 1- AGACCAGTTT lnaAs;omeGs;lnaAs;omeCs;lnaCs;omeAs;lnaGs;omeUs;lnaTs;omeUs; lnaT 3650 9 mOl TACCT s;omeAs;lnaCs;omeCs;lnaT-Sup SM N l- CCTAGCTACT lnaCs;omeCs;lnaTs;omeAs;lnaGs;omeCs;lnaTs;omeAs;lnaCs;omeUs; lnaT 13093 60 mOl TTGAA s;omeUs;lnaGs;omeAs;lnaA-Sup

SM N 1- TCCTAGCTAC lnaTs;omeCs;lnaCs;omeUs;lnaAs;omeGs;lnaCs;omeUs;lnaAs;omeCs; lnaT 13094 1 mOl TTTGA s;omeUs;lnaTs;omeGs;lnaA-Sup

SM N 1- GAAATATTCC lnaGs;omeAs;lnaAs;omeAs;lnaTs;omeAs;lnaTs;omeUs;lnaCs;omeCs; lnaT 10065 2 mOl TTATA s;omeUs;lnaAs;omeUs;lnaA-Sup

SM N 1- AAATATTCCT lnaAs;omeAs;lnaAs;omeUs;lnaAs;omeUs;lnaTs;omeCs;lnaCs;omeUs; lnaT 10066 3 mOl TATAG s;omeAs;lnaTs;omeAs;lnaG-Sup

SM N 1- AATATTCCTT lnaAs;omeAs;lnaTs;omeAs;lnaTs;omeUs;lnaCs;omeCs;lnaTs;omeUs; lnaA 10067 4 mOl ATAGC s;omeUs;lnaAs;omeGs;lnaC-Sup

SM N 1- ATATTCCTTA lnaAs;omeUs;lnaAs;omeUs;lnaTs;omeCs;lnaCs;omeUs;lnaTs;omeAs; lnaT 10068 5 mOl TAGCC s;omeAs;lnaGs;omeCs;lnaC-Sup

SM N 1- TATTCCTTAT lnaTs;omeAs;lnaTs;omeUs;lnaCs;omeCs;lnaTs;omeUs;lnaAs;omeUs; lnaA 10069 6 mOl AGCCA s;omeGs;lnaCs;omeCs;lnaA-Sup

SM N 1- ATTCCTTATA lnaAs;omeUs;lnaTs;omeCs;lnaCs;omeUs;lnaTs;omeAs;lnaTs;omeAs; lnaG 10070 7 mOl GCCAG s;omeCs;lnaCs;omeAs;lnaG-Sup

SM N 1- TTCCTTATAG lnaTs;omeUs;lnaCs;omeCs;lnaTs;omeUs;lnaAs;omeUs;lnaAs;omeGs; lnaC 10071 8 mOl CCAGG s;omeCs;lnaAs;omeGs;lnaG-Sup

SM N 1- TCCTT ATAGC lnaTs;omeCs;lnaCs;omeUs;lnaTs;omeAs;lnaTs;omeAs;lnaGs;omeCs; lnaC 10072 9 mOl CAGGT s;omeAs;lnaGs;omeGs;lnaT-Sup

SM N 1- CCTTATAGCC lnaCs;omeCs;lnaTs;omeUs;lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeCs; lnaA 10073 0 mOl AGGTC s;omeGs;lnaGs;omeUs;lnaC-Sup

SM N 1- CTTATAGCCA lnaCs;omeUs;lnaTs;omeAs;lnaTs;omeAs;lnaGs;omeCs;lnaCs;omeAs; lnaG 10074 1 mOl GGTCT s;omeGs;lnaTs;omeCs;lnaT-Sup

SM N 1- TTATAGCCAG lnaTs;omeUs;lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeCs;lnaAs;omeGs; lna 10075 2 mOl GTCTA Gs;omeUs;lnaCs;omeUs;lnaA-Sup

SM N 1- GCCAG GTCTA lnaGs;omeCs;lnaCs;omeAs;lnaGs;omeGs;lnaTs;omeCs;lnaTs;omeAs; lnaA 10080 3 mOl AAATT s;omeAs;lnaAs;omeUs;lnaT-Sup

SM N 1- CCAGGTCTAA lnaCs;omeCs;lnaAs;omeGs;lnaGs;omeUs;lnaCs;omeUs;lnaAs;omeAs; lna 10081 4 mOl AATTC As;omeAs;lnaTs;omeUs;lnaC-Sup

SM N 1- CAGGTCTAAA lnaCs;omeAs;lnaGs;omeGs;lnaTs;omeCs;lnaTs;omeAs;lnaAs;omeAs; lnaA 10082 5 mOl ATTCA s;omeUs;lnaTs;omeCs;lnaA-Sup

SM N 1- GGTCTAAAAT lnaGs;omeGs;lnaTs;omeCs;lnaTs;omeAs;lnaAs;omeAs;lnaAs;omeUs; lnaT 10084 6 m01 TCAAT s;omeCs;lnaAs;omeAs;lnaT-Sup

SM N 1- CTAAAATTCA lnaCs;omeUs;lnaAs;omeAs;lnaAs;omeAs;lnaTs;omeUs;lnaCs;omeAs; lnaA 10087 7 mOl ATGGC s;omeUs;lnaGs;omeGs;lnaC-Sup

SM N 1- CTAAAATTCA omeCs;omeUs;omeAs;omeAs;omeAs;omeAs;omeUs;omeUs;omeCs;ome 10087 7 m03 ATGGC As;omeAs;omeUs;omeGs;omeGs;omeC-Sup

SM N 1- GGACCACCA lnaGs;omeGs;lnaAs;omeCs;lnaCs;omeAs;lnaCs;omeCs;lnaAs;omeGs; lnaT 10168 8 mOl GTAAGT s;omeAs;lnaAs;omeGs;lnaT-Sup

SM N 1- GACCACCAGT dGs;lnaAs;dCs;lnaCs;dAs;lnaCs;dCs;lnaAs;dGs;lnaTs;dAs;lnaAs; dGs;lnaTs; 10169 9 m02 AAGTA dA-Sup

SM N 1- GACCACCAGT lnaGs;omeAs;lnaCs;omeCs;lnaAs;omeCs;lnaCs;omeAs;lnaGs;omeUs; lnaA 10169 9 mOl AAGTA s;omeAs;lnaGs;omeUs;lnaA-Sup

SM N 1- ACCACCAGTA dAs;lnaCs;dCs;lnaAs;dCs;lnaCs;dAs;lnaGs;dTs;lnaAs;dAs;lnaGs; dTs;lnaAs; 10170 0 m02 AGTAA dA-Sup

SM N 1- ACCACCAGTA lnaAs;omeCs;lnaCs;omeAs;lnaCs;omeCs;lnaAs;omeGs;lnaTs;omeAs; lnaA 10170 0 mOl AGTAA s;omeGs;lnaTs;omeAs;lnaA-Sup

SM N 1- TTCTGTTACC lnaTs;omeUs;lnaCs;omeUs;lnaGs;omeUs;lnaTs;omeAs;lnaCs;omeCs; lnaC 10337 1 mOl CAGAT s;omeAs;lnaGs;omeAs;lnaT-Sup

SM N 1- TCTGTTACCC lnaTs;omeCs;lnaTs;omeGs;lnaTs;omeUs;lnaAs;omeCs;lnaCs;omeCs; lnaA 10338 2 mOl AGATG s;omeGs;lnaAs;omeUs;lnaG-Sup SM N l- CTGTTACCCA lnaCs;omeUs;lnaGs;omeUs;lnaTs;omeAs;lnaCs;omeCs;lnaCs;omeAs; lnaG 10339 83 mOl GATGC s;omeAs;lnaTs;omeGs;lnaC-Sup

SM N 1- 1 1 1 1 I AG I A dTs;lnaTs;dTs;lnaTs;dTs;lnaAs;dGs;lnaGs;dTs;lnaAs;dTs;lnaTs; dAs;lnaAs; 10763 84 m02 TTAAC dC-Sup

SM N 1- 1 1 1 1 I AG I A lnaTs;omeUs;lnaTs;omeUs;lnaTs;omeAs;lnaGs;omeGs;lnaTs;omeAs; lnaT 10763 84 mOl TTAAC s;omeUs;lnaAs;omeAs;lnaC-Sup

SM N 1- 1 1 1 I AG I A I lnaTs;omeUs;lnaTs;omeUs;lnaAs;omeGs;lnaGs;omeUs;lnaAs;omeUs; lna 10764 85 mOl TAACA Ts;omeAs;lnaAs;omeCs;lnaA-Sup

SM N 1- CATAGCTTCA lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeCs;lnaTs;omeUs;lnaCs;omeAs; lnaT 10949 86 mOl TAGTG s;omeAs;lnaGs;omeUs;lnaG-Sup

SM N 1- TAGCTTCATA lnaTs;omeAs;lnaGs;omeCs;lnaTs;omeUs;lnaCs;omeAs;lnaTs;omeAs; lnaG 10951 87 mOl GTGGA s;omeUs;lnaGs;omeGs;lnaA-Sup

SM N 1- AGCTTCATAG lnaAs;omeGs;lnaCs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs; lnaT 10952 88 mOl TGGAA s;omeGs;lnaGs;omeAs;lnaA-Sup

SM N 1- GCTTCATAGT lnaGs;omeCs;lnaTs;omeUs;lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeUs; lnaG 10953 89 mOl GGAAC s;omeGs;lnaAs;omeAs;lnaC-Sup

SM N 1- CTTCATAGTG lnaCs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs; lnaG 10954 90 mOl GAACA s;omeAs;lnaAs;omeCs;lnaA-Sup

SM N 1- TCATGGTACA lnaTs;omeCs;lnaAs;omeUs;lnaGs;omeGs;lnaTs;omeAs;lnaCs;omeAs; lnaT 11415 91 mOl TGAGT s;omeGs;lnaAs;omeGs;lnaT-Sup

SM N 1- TGGTACATGA lnaTs;omeGs;lnaGs;omeUs;lnaAs;omeCs;lnaAs;omeUs;lnaGs;omeAs; lna 11418 92 mOl GTGGC Gs;omeUs;lnaGs;omeGs;lnaC-Sup

SM N 1- GGTACATGA dGs;lnaGs;dTs;lnaAs;dCs;lnaAs;dTs;lnaGs;dAs;lnaGs;dTs;lnaGs; dGs;lnaCs 11419 93 m02 GTGGCT ;dT-Sup

SM N 1- GGTACATGA lnaGs;omeGs;lnaTs;omeAs;lnaCs;omeAs;lnaTs;omeGs;lnaAs;omeGs; lnaT 11419 93 mOl GTGGCT s;omeGs;lnaGs;omeCs;lnaT-Sup

SM N 1- TACATGAGTG lnaTs;omeAs;lnaCs;omeAs;lnaTs;omeGs;lnaAs;omeGs;lnaTs;omeGs; lnaG 11421 94 mOl GCTAT s;omeCs;lnaTs;omeAs;lnaT-Sup

SM N 1- ACATGAGTG lnaAs;omeCs;lnaAs;omeUs;lnaGs;omeAs;lnaGs;omeUs;lnaGs;omeGs; lna 11422 95 mOl GCTATC Cs;omeUs;lnaAs;omeUs;lnaC-Sup

SM N 1- CATGAGTGG lnaCs;omeAs;lnaTs;omeGs;lnaAs;omeGs;lnaTs;omeGs;lnaGs;omeCs; lnaT 11423 96 m01 CTATCA s;omeAs;lnaTs;omeCs;lnaA-Sup

SM N 1- CTGGCTATTA lnaCs;omeUs;lnaGs;omeGs;lnaCs;omeUs;lnaAs;omeUs;lnaTs;omeAs; lnaT 11440 97 mOl TATGG s;omeAs;lnaTs;omeGs;lnaG-Sup

SM N 1- TGGCTATTAT lnaTs;omeGs;lnaGs;omeCs;lnaTs;omeAs;lnaTs;omeUs;lnaAs;omeUs; lnaA 11441 98 mOl ATGGT s;omeUs;lnaGs;omeGs;lnaT-Sup

SM N 1- GGCTATTATA lnaGs;omeGs;lnaCs;omeUs;lnaAs;omeUs;lnaTs;omeAs;lnaTs;omeAs; lnaT 11442 99 mOl TGGTA s;omeGs;lnaGs;omeUs;lnaA-Sup

SM N 1- GCTATTATAT lnaGs;omeCs;lnaTs;omeAs;lnaTs;omeUs;lnaAs;omeUs;lnaAs;omeUs; lna 11443 100 GGTAA Gs;omeGs;lnaTs;omeAs;lnaA-Sup

mOl

SM N 1- GTATCATCTG lnaGs;omeUs;lnaAs;omeUs;lnaCs;omeAs;lnaTs;omeCs;lnaTs;omeGs; lnaT 12369 101 TGTGT s;omeGs;lnaTs;omeGs;lnaT-Sup

mOl

SM N 1- GCTTTGGGA lnaGs;omeCs;lnaTs;omeUs;lnaTs;omeGs;lnaGs;omeGs;lnaAs;omeAs; lna 13097 102 AGTATG 1 1 1 1 Gs;omeUs;lnaAs;omeUs;lnaG;dT;dT;dT;dT;lnaTs;omeCs;lnaAs;omeC s;lna mOl TCACTTTCAT Ts;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeAs;lnaTs;omeGs;lna Cs;o

AATGCTGG meUs;lnaGs;omeG-Sup

SM N 1- CTTTGGGAA lnaCs;omeUs;lnaTs;omeUs;lnaGs;omeGs;lnaGs;omeAs;lnaAs;omeGs; lna 13102 103 GTATG 1 1 1 1 1 Ts;omeAs;lnaTs;omeGs;lnaT;dT;dT;dT;dT;lnaTs;omeCs;lnaAs;omeC s;lnaT mOl TCACTTTCAT s;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeAs;lnaTs;omeGs;lnaC s;o

AATGCTGG meUs;lnaGs;omeG-Sup SM N l- GGTACATGA lnaGs;omeGs;lnaTs;omeAs;lnaCs;omeAs;lnaTs;omeGs;lnaAs;omeGs; lnaT 13099 104 GTGGC I 1 1 1 1 s;omeGs;lnaGs;omeCs;lnaT;dT;dT;dT;dT;lnaTs;omeCs;lnaAs;omeCs ;lnaTs mOl TCACTTTCAT ;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeAs;lnaTs;omeGs;lnaCs ;om

AATGCTGG eUs;lnaGs;omeG-Sup

SM N 1- TGATGCTGAT lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeGs;lnaAs;omeUs; lna 13103 105 GC I 1 1 1 1 1 I U Gs;omeCs;lnaTs;omeUs;lnaT;dT;dT;dT;dT;lnaCs;omeUs;lnaAs;omeA s;lna mOl AAAATTCAAT As;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lna C- GGC Sup

SM N 1- CTAAAATTCA lnaCs;omeUs;lnaAs;omeAs;lnaAs;omeAs;lnaTs;omeUs;lnaCs;omeAs; lnaA 13104 106 ATGGC I I M C s;omeUs;lnaGs;omeGs;lnaC;dT;dT;dT;dT;lnaCs;omeUs;lnaAs;omeAs ;lnaA mOl TAAAATTCAA s;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lnaC - TGGC Sup

SM N 1- CTGTTACCCA lnaCs;omeUs;lnaGs;omeUs;lnaTs;omeAs;lnaCs;omeCs;lnaCs;omeAs; lnaG 13105 107 GATGC 1 1 1 1 C s;omeAs;lnaTs;omeGs;lnaC;dT;dT;dT;dT;lnaCs;omeUs;lnaAs;omeAs ;lnaA mOl TAAAATTCAA s;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lnaC - TGGC Sup

SM N 1- CTTCATAGTG lnaCs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs; lnaG 13106 108 GAACA 1 1 1 1 C s;omeAs;lnaAs;omeCs;lnaA;dT;dT;dT;dT;lnaCs;omeUs;lnaAs;omeAs ;lnaA mOl TAAAATTCAA s;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lnaC - TGGC Sup

SM N 1- TCACTTTCAT lnaTs;omeCs;lnaAs;omeCs;lnaTs;omeUs;lnaTs;omeCs;lnaAs;omeUs; lnaA 13107 109 AATGCTGGTT s;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeG;dT;dT;dT;dT;lnaTs ;ome mOl TTTCACTTTC Cs;lnaAs;omeCs;lnaTs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;ome As;ln

ATAATGCTGG aTs;omeGs;lnaCs;omeUs;lnaGs;omeG-Sup

SM N l- AAATTCAATG lnaAs;omeAs;lnaAs;omeUs;lnaTs;omeCs;lnaAs;omeAs;lnaTs;omeGs; lnaG 10090 110 GCCCA s;omeCs;lnaCs;omeCs;lnaA-Sup

mOl

SM N 1- AATTCAATGG lnaAs;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs; lna 10091 111 CCCAC Cs;omeCs;lnaCs;omeAs;lnaC-Sup

mOl

SM N 1- ATTCAATGGC lnaAs;omeUs;lnaTs;omeCs;lnaAs;omeAs;lnaTs;omeGs;lnaGs;omeCs; lnaC 10092 112 CCACC s;omeCs;lnaAs;omeCs;lnaC-Sup

mOl

SM N 1- TTCAATGGCC lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lnaCs;omeCs; lnaC 10093 113 CACCA s;omeAs;lnaCs;omeCs;lnaA-Sup

mOl

SM N 1- TCAATGGCCC lnaTs;omeCs;lnaAs;omeAs;lnaTs;omeGs;lnaGs;omeCs;lnaCs;omeCs; lnaA 10094 114 ACCAC s;omeCs;lnaCs;omeAs;lnaC-Sup

mOl

SM N 1- CAATG GCCCA lnaCs;omeAs;lnaAs;omeUs;lnaGs;omeGs;lnaCs;omeCs;lnaCs;omeAs; lnaC 10095 115 CCACC s;omeCs;lnaAs;omeCs;lnaC-Sup

mOl

SM N 1- AATGGCCCAC lnaAs;omeAs;lnaTs;omeGs;lnaGs;omeCs;lnaCs;omeCs;lnaAs;omeCs; lnaC 10096 116 CACCG s;omeAs;lnaCs;omeCs;lnaG-Sup

mOl

SM N 1- ATGGCCCACC lnaAs;omeUs;lnaGs;omeGs;lnaCs;omeCs;lnaCs;omeAs;lnaCs;omeCs; lnaA 10097 117 ACCGC s;omeCs;lnaCs;omeGs;lnaC-Sup

mOl

SM N 1- AATGCCTTTC lnaAs;omeAs;lnaTs;omeGs;lnaCs;omeCs;lnaTs;omeUs;lnaTs;omeCs; lnaTs 10330 118 TGTTA ;omeGs;lnaTs;omeUs;lnaA-Sup

mOl SM N l- ATGCCTTTCT lnaAs;omeUs;lnaGs;omeLs;lnaLs;omeUs;lnaTs;omeUs;lnaLs;omeUs; lna 10331 119 GTTAC Gs;omeUs;lnaTs;omeAs;lnaL-Sup

mOl

SM N 1- TGCCTTTCTG lnaTs;omeGs;lnaLs;omeLs;lnaTs;omeUs;lnaTs;omeLs;lnaTs;omeGs; lnaTs 10332 120 TTACC ;omeUs;lnaAs;omeLs;lnaL-Sup

mOl

SM N 1- GCCTTTCTGT lnaGs;omeLs;lnaLs;omeUs;lnaTs;omeUs;lnaLs;omeUs;lnaGs;omeUs; lnaT 10333 121 TACCC s;omeAs;lnaLs;omeLs;lnaL-Sup

mOl

SM N 1- CCTTTCTGTT lnaLs;omeLs;lnaTs;omeUs;lnaTs;omeLs;lnaTs;omeGs;lnaTs;omeUs; lnaA 10334 122 ACCCA s;omeLs;lnaLs;omeLs;lnaA-Sup

mOl

SM N 1- CTTTCTGTTA lnaLs;omeUs;lnaTs;omeUs;lnaLs;omeUs;lnaGs;omeUs;lnaTs;omeAs; lnaL 10335 123 CCCAG s;omeLs;lnaLs;omeAs;lnaG-Sup

mOl

SM N 1- TTTCTGTTAC lnaTs;omeUs;lnaTs;omeLs;lnaTs;omeGs;lnaTs;omeUs;lnaAs;omeLs; lnaL 10336 124 CCAGA s;omeLs;lnaAs;omeGs;lnaA-Sup

mOl

SM N 1- TGTTACCCAG lnaTs;omeGs;lnaTs;omeUs;lnaAs;omeLs;lnaLs;omeLs;lnaAs;omeGs; lnaA 10340 125 ATGCA s;omeUs;lnaGs;omeLs;lnaA-Sup

mOl

SM N 1- GTTACCCAGA lnaGs;omeUs;lnaTs;omeAs;lnaLs;omeLs;lnaLs;omeAs;lnaGs;omeAs; lnaT 10341 126 TGCAG s;omeGs;lnaLs;omeAs;lnaG-Sup

mOl

SM N 1- TTACCCAGAT lnaTs;omeUs;lnaAs;omeLs;lnaLs;omeLs;lnaAs;omeGs;lnaAs;omeUs; lnaG 10342 127 GCAGT s;omeLs;lnaAs;omeGs;lnaT-Sup

mOl

SM N 1- ACCCAGATGC lnaAs;omeLs;lnaLs;omeLs;lnaAs;omeGs;lnaAs;omeUs;lnaGs;omeLs; lnaA 10344 128 AGTGC s;omeGs;lnaTs;omeGs;lnaL-Sup

mOl

SM N 1- CCCAGATGCA lnaLs;omeLs;lnaLs;omeAs;lnaGs;omeAs;lnaTs;omeGs;lnaLs;omeAs; lnaG 10345 129 GTGCT s;omeUs;lnaGs;omeLs;lnaT-Sup

mOl

SM N 1- CCAGATGCA lnaLs;omeLs;lnaAs;omeGs;lnaAs;omeUs;lnaGs;omeLs;lnaAs;omeGs; lnaT 10346 130 GTGCTC s;omeGs;lnaLs;omeUs;lnaL-Sup

mOl

SM N 1- CAGATGCAGT lnaLs;omeAs;lnaGs;omeAs;lnaTs;omeGs;lnaLs;omeAs;lnaGs;omeUs; lna 10347 131 GCTCT Gs;omeLs;lnaTs;omeLs;lnaT-Sup

mOl

SM N 1- AGATGCAGT lnaAs;omeGs;lnaAs;omeUs;lnaGs;omeLs;lnaAs;omeGs;lnaTs;omeGs; lna 10348 132 GCTCTT Ls;omeUs;lnaLs;omeUs;lnaT-Sup

mOl

SM N 1- 1 1 1 1 AC 1 LA 1 lnaTs;omeUs;lnaTs;omeUs;lnaAs;omeLs;lnaTs;omeLs;lnaAs;omeUs; lnaA 10942 133 AG TT s;omeGs;lnaLs;omeUs;lnaT-Sup

mOl

SM N 1- TTTALTLATA lnaTs;omeUs;lnaTs;omeAs;lnaLs;omeUs;lnaLs;omeAs;lnaTs;omeAs; lnaG 10943 134 GLTTL s;omeLs;lnaTs;omeUs;lnaL-Sup

mOl

SM N 1- TTALTLATAG lnaTs;omeUs;lnaAs;omeLs;lnaTs;omeLs;lnaAs;omeUs;lnaAs;omeGs; lnaL 10944 135 LTTLA s;omeUs;lnaTs;omeLs;lnaA-Sup

mOl SM N 1- TACTCATAGC lnaTs;omeAs;lnaCs;omeUs;lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeCs; lnaT 10945 136 TTCAT s;omeUs;lnaCs;omeAs;lnaT-Sup

mOl

SM N 1- ACTCATAGCT lnaAs;omeCs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeUs; lnaT 10946 137 TCATA s;omeCs;lnaAs;omeUs;lnaA-Sup

mOl

SM N 1- CTCATAGCTT lnaCs;omeUs;lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeCs;lnaTs;omeUs; lnaC 10947 138 CATAG s;omeAs;lnaTs;omeAs;lnaG-Sup

mOl

SM N 1- TCATAGCTTC lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeUs;lnaTs;omeCs; lnaA 10948 139 ATAGT s;omeUs;lnaAs;omeGs;lnaT-Sup

mOl

SM N 1- ATAGCTTCAT lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeUs;lnaTs;omeCs;lnaAs;omeUs; lna 10950 140 AGTGG As;omeGs;lnaTs;omeGs;lnaG-Sup

mOl

SM N 1- TTCATAGTGG lnaTs;omeUs;lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeUs;lnaGs;omeGs; lna 10955 141 AACAG As;omeAs;lnaCs;omeAs;lnaG-Sup

mOl

SM N 1- TCATAGTGGA lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs;lnaGs;omeAs; lnaA 10956 142 ACAGA s;omeCs;lnaAs;omeGs;lnaA-Sup

mOl

SM N 1- CATAGTGGA lnaCs;omeAs;lnaTs;omeAs;lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs; lnaC 10957 143 ACAGAT s;omeAs;lnaGs;omeAs;lnaT-Sup

mOl

SM N 1- ATAGTGGAA lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs;lnaGs;omeAs;lnaAs;omeCs; lna 10958 144 CAGATA As;omeGs;lnaAs;omeUs;lnaA-Sup

mOl

SM N 1- TAGTGGAAC lnaTs;omeAs;lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs;lnaCs;omeAs; lna 10959 145 AGATAC Gs;omeAs;lnaTs;omeAs;lnaC-Sup

mOl

SM N 1- AGTGGAACA lnaAs;omeGs;lnaTs;omeGs;lnaGs;omeAs;lnaAs;omeCs;lnaAs;omeGs; lna 10960 146 GATACA As;omeUs;lnaAs;omeCs;lnaA-Sup

mOl

SM N 1- GTGGAACAG lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs;lnaCs;omeAs;lnaGs;omeAs; lna 10961 147 ATACAT Ts;omeAs;lnaCs;omeAs;lnaT-Sup

mOl

SM N 1- TGGAACAGA lnaTs;omeGs;lnaGs;omeAs;lnaAs;omeCs;lnaAs;omeGs;lnaAs;omeUs; lna 10962 148 TACATA As;omeCs;lnaAs;omeUs;lnaA-Sup

mOl

SM N 1- TGTCCAGATT lnaTs;omeGs;lnaTs;omeCs;lnaCs;omeAs;lnaGs;omeAs;lnaTs;omeUs; lnaC 11367 149 CTCTT s;omeUs;lnaCs;omeUs;lnaT-Sup

mOl

SM N 1- GTCCAGATTC lnaGs;omeUs;lnaCs;omeCs;lnaAs;omeGs;lnaAs;omeUs;lnaTs;omeCs; lnaT 11368 150 TCTTG s;omeCs;lnaTs;omeUs;lnaG-Sup

mOl

SM N 1- TCCAGATTCT lnaTs;omeCs;lnaCs;omeAs;lnaGs;omeAs;lnaTs;omeUs;lnaCs;omeUs; lnaC 11369 151 CTTGA s;omeUs;lnaTs;omeGs;lnaA-Sup

mOl

SM N 1- CCAGATTCTC lnaCs;omeCs;lnaAs;omeGs;lnaAs;omeUs;lnaTs;omeCs;lnaTs;omeCs; lnaT 11370 152 TTGAT s;omeUs;lnaGs;omeAs;lnaT-Sup

mOl - I l l -

SM N 1- CAGATTCTCT lnaCs;omeAs;lnaGs;omeAs;lnaTs;omeUs;lnaCs;omeUs;lnaCs;omeUs; lnaT 11371 153 TGATG s;omeGs;lnaAs;omeUs;lnaG-Sup

mOl

SM N 1- AGATTCTCTT lnaAs;omeGs;lnaAs;omeUs;lnaTs;omeCs;lnaTs;omeCs;lnaTs;omeUs; lnaG 11372 154 GATGA s;omeAs;lnaTs;omeGs;lnaA-Sup

mOl

SM N 1- GATTCTCTTG lnaGs;omeAs;lnaTs;omeUs;lnaCs;omeUs;lnaCs;omeUs;lnaTs;omeGs; lnaA 11373 155 ATGAT s;omeUs;lnaGs;omeAs;lnaT-Sup

mOl

SM N 1- GGAAGTATG lnaGs;omeGs;lnaAs;omeAs;lnaGs;omeUs;lnaAs;omeUs;lnaGs;omeUs; lna 11400 156 TTAATT Ts;omeAs;lnaAs;omeUs;lnaT-Sup

mOl

SM N 1- GAAGTATGTT lnaGs;omeAs;lnaAs;omeGs;lnaTs;omeAs;lnaTs;omeGs;lnaTs;omeUs; lnaA 11401 157 AATTT s;omeAs;lnaTs;omeUs;lnaT-Sup

mOl

SM N 1- AAGTATGTTA lnaAs;omeAs;lnaGs;omeUs;lnaAs;omeUs;lnaGs;omeUs;lnaTs;omeAs; lna 11402 158 ATTTC As;omeUs;lnaTs;omeUs;lnaC-Sup

mOl

SM N 1- AGTATGTTAA lnaAs;omeGs;lnaTs;omeAs;lnaTs;omeGs;lnaTs;omeUs;lnaAs;omeAs; lnaT 11403 159 TTTCA s;omeUs;lnaTs;omeCs;lnaA-Sup

mOl

SM N 1- GTATGTTAAT lnaGs;omeUs;lnaAs;omeUs;lnaGs;omeUs;lnaTs;omeAs;lnaAs;omeUs; lna 11404 160 TTCAT Ts;omeUs;lnaCs;omeAs;lnaT-Sup

mOl

SM N 1- TATG TTAATT lnaTs;omeAs;lnaTs;omeGs;lnaTs;omeUs;lnaAs;omeAs;lnaTs;omeUs; lnaT 11405 161 TCATG s;omeCs;lnaAs;omeUs;lnaG-Sup

mOl

SM N 1- ATGTTAATTT lnaAs;omeUs;lnaGs;omeUs;lnaTs;omeAs;lnaAs;omeUs;lnaTs;omeUs; lna 11406 162 CATGG Cs;omeAs;lnaTs;omeGs;lnaG-Sup

mOl

SM N 1- TGAAATATTC lnaTs;omeGs;lnaAs;omeAs;lnaAs;omeUs;lnaAs;omeUs;lnaTs;omeCs; lnaC 10064 163 CTTAT s;omeUs;lnaTs;omeAs;lnaT-Sup

mOl

SM N 1- TATAGCCAGG lnaTs;omeAs;lnaTs;omeAs;lnaGs;omeCs;lnaCs;omeAs;lnaGs;omeGs; lnaT 10076 164 TCTAA s;omeCs;lnaTs;omeAs;lnaA-Sup

mOl

SM N 1- ATAGCCAGGT lnaAs;omeUs;lnaAs;omeGs;lnaCs;omeCs;lnaAs;omeGs;lnaGs;omeUs; lna 10077 165 CTAAA Cs;omeUs;lnaAs;omeAs;lnaA-Sup

mOl

SM N 1- AGGTCTAAAA lnaAs;omeGs;lnaGs;omeUs;lnaCs;omeUs;lnaAs;omeAs;lnaAs;omeAs; lna 10083 166 TTCAA Ts;omeUs;lnaCs;omeAs;lnaA-Sup

mOl

SM N 1- GTCTAAAATT lnaGs;omeUs;lnaCs;omeUs;lnaAs;omeAs;lnaAs;omeAs;lnaTs;omeUs; lna 10085 167 CAATG Cs;omeAs;lnaAs;omeUs;lnaG-Sup

mOl

SM N 1- TCTAAAATTC lnaTs;omeCs;lnaTs;omeAs;lnaAs;omeAs;lnaAs;omeUs;lnaTs;omeCs; lnaA 10086 168 AATGG s;omeAs;lnaTs;omeGs;lnaG-Sup

mOl

SM N 1- TAAAATTCAA lnaTs;omeAs;lnaAs;omeAs;lnaAs;omeUs;lnaTs;omeCs;lnaAs;omeAs; lnaT 10088 169 TGGCC s;omeGs;lnaGs;omeCs;lnaC-Sup

mOl SMNl- AAAATTCAAT lnaAs;omeAs;lnaAs;omeAs;lnaTs;omeUs;lnaCs;omeAs;lnaAs;omeUs; lnaG 10089

170 GGCCC s;omeGs;lnaCs;omeCs;lnaC-Sup

mOl

unc-232 CTACGCGTCG lnaCs;dTs;lnaAs;dCs;lnaGs;dCs;lnaGs;dTs;lnaCs;dGs;lnaAs;dCs; lnaGs;dGs; 13095 ml2 ACGGT InaT-Sup

unc-232 CTACGCGTCG lnaCs;omeUs;lnaAs;omeCs;lnaGs;omeCs;lnaGs;omeUs;lnaCs;omeGs; lna 13095 mOl ACGGT As;omeCs;lnaGs;omeGs;lnaT-Sup

unc-293 CCGATTCGCG lnaCs;dCs;lnaGs;dAs;lnaTs;dTs;lnaCs;dGs;lnaCs;dGs;lnaCs;dGs; lnaTs;dAs; 13096 ml2 CGTAA InaA-Sup

unc-293 CCGATTCGCG lnaCs;omeCs;lnaGs;omeAs;lnaTs;omeUs;lnaCs;omeGs;lnaCs;omeGs; lnaC 13096 mOl CGTAA s;omeGs;lnaTs;omeAs;lnaA-Sup

SMN1- TGATGCTGA lnaTs;omeGs;lnaAs;omeUs;lnaGs;omeCs;lnaTs;omeGs;lnaAs;ome 13108

246 TGCT Us;lnaGs;omeCs;lnaT

mOl

SMN1- TGATGCTGA omeUs;lnaGs;omeAs;lnaTs;omeGs;lnaCs;omeUs;lnaGs;omeAs;lna 13108

246 TGCT Ts;omeGs;lnaCs;omeU

ml6

SMN1- TGATGCTGA lnaTs;omeGs;omeAs;lnaTs;omeGs;omeCs;lnaTs;omeGs;omeAs;ln 13109

37 ml9 TGCTTT aTs;omeGs;omeCs;lnaTs;omeUs;omeU

SMN1- TGATGCTGA lnaTs;lnaGs;omeAs;omeUs;lnaGs;lnaCs;omeUs;omeGs;lnaAs;lnaT 13109

37 m20 TGCTTT s;omeGs;omeCs;lnaTs;lnaTs;omeU

SMN1- CTTCATAGT lnaCs;omeUs;lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;ome 13110

435 GG Gs;lnaG

mOl

SMN1- TCATAGTGG lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs;lnaGs;ome 13111

438 AACA As;lnaAs;omeCs;lnaA

mOl

SMN1- TCATAGTGG lnaTs;omeCs;lnaAs;omeUs;lnaAs;omeGs;lnaTs;omeGs;lnaGs;ome 13112

439 AA As;lnaA

mOl

SMN1- CTTCATAGT lnaCs;lnaTs;omeUs;lnaCs;lnaAs;omeUs;lnaAs;lnaGs;omeUs;lnaGs; l 10954

90 ml8 GGAACA naGs;omeAs;lnaAs;lnaCs;omeA

SMN1- CTTCATAGT lnaCs;omeUs;omeUs;lnaCs;omeAs;omeUs;lnaAs;omeGs;omeUs;ln 10954

90 ml9 GGAACA aGs;omeGs;omeAs;lnaAs;omeCs;omeA

SMN1- CTTCATAGT lnaCs;lnaTs;omeUs;omeCs;lnaAs;lnaTs;omeAs;omeGs;lnaTs;lnaGs 10954

90 m20 GGAACA ;omeGs;omeAs;lnaAs;lnaCs;omeA

SMN1- GTGGAACA lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs;lnaCs;omeAs;lnaGs;ome 13113

457 GATAC As;lnaTs;omeAs;lnaC

mOl

SMN1- GTGGAACA lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs;lnaCs;omeAs;lnaGs;ome 13114

458 GAT As;lnaT

mOl

SMN1- GTGGAACA lnaGs;omeUs;lnaGs;omeGs;lnaAs;omeAs;lnaCs;omeAs;lnaG 13115

459 G

mOl

Example 3. Confirmation ofPRC2 mechanism of action MATERIALS AND METHODS:

Oligonucleotides

The oligos used in Example 3 are shown in Table 6 below and/or in Table 4 (See Table 3 for structural features of formatted sequence).

Table 6.

Non Human Primate (NHP) dose response curves

Non-human primate Cynomolgous monkey fibroblasts (NHP) was purchased from the Coriell Institute (Coriell cat#: AG21329) and cultured in MEM (Eagle) Alpha modification with nucleosides and 2mM L-Glutamine and 15% FBS. Cells were plated at 1 x 10 4 cells per well of 96 well plate. 24 hours later oligonucleotides were transfected into cells with

Lipofectamine 2000 (LifeTechnology cat# 11668-019) generating a dose response curve spanning 200 - 1.5nM final concentrations. Oligos combined with cells without media change for 48 hours prior to mRNA expression analysis. qPCR

For SMN full length (FL) qPCR, cDNA was prepared using Cells to Ct

(LifeTechnology cat# 4391851C and 4391852C) according to manufacturer's instructions. qPCR was performed using TaqMan probes (IDT) designed previously (Hua et al 2010 Genes and Dev 24: 1634-1644) and housekeeping GAPDH TaqMan assay (LifeTechnology cat # Hs02758991_gl) Analysis was conducted by AACt method.

Full Length SMN probes:

Fwd primer: 5' -GCTGATGCTTTGGGAAGTATGTTA-3' (SEQ ID NO: 13117) Rev primer: 5 ' -C ACCTTCCTTCTTTTTGATTTTGTC-3 ' (SEQ ID NO: 13118) Probe 5 ' -TACATGAGTGGCTATCATACT-3 ' (SEQ ID NO: 13119)

For SMN full length (FL) and Δ7 (del7) SMN qPCR, cDNA was prepared using Cells to Ct (LifeTechnology cat# 4391851C and 4391852C) according to manufacturer's instructions. qPCR was performed using TaqMan probes (IDT) designed previously (Hua et al 2010 Genes and Dev 24: 1634-1644) and housekeeping GAPDH TaqMan assay

(LifeTechnology cat # Hs02758991_gl). Analysis was conducted by AACt method.

Full Length SMN probes:

Fwd primer: 5 '-GCTGATGCTTTGGGAAGTATGTTA-3' (SEQ ID NO: 13117) Rev primer: 5 '-C ACCTTCCTTCTTTTTGATTTTGTC-3' (SEQ ID NO: 13118) Probe 5 '-TACATGAGTGGCTATCATACT-3' (SEQ ID NO: 13119)

Δ7 SMN probe:

Fwd Primer: 5'-TGGACCACCAATAATTCCCC-3' (SEQ ID NO: 13120)

Rev Primer: 5 ' -ATGCCAGCATTTCCATATAATAGCC-3 ' (SEQ ID NO: 13121)

Probe 5'-TCCAGATTCTCTTGATGATG-3' (SEQ ID NO: 13122) PRC2-associated SMN Oligos in combination with splice-switching oligo experiments

To assess effects of PRC2-associated SMN oligos with a splice- switching oligo a matrixed approach consisting of two different methods was taken, (i) While maintaining the concentration of PRC2 SMN oligo at 30nM, the splice- switching oligo was combined at the following concentrations of 192, 96, 48, 24, 12, 6, 3 and 1.5pM. (ii) While maintaining the concentration of splice- switching oligo at 300pM, the PRC2 SMN oligo was combined at the following concentrations of 100, 50, 25, 12.5, 6.25, 3.125, 1.56nM. For each set of experiments these combinations were transfected into SMA derived fibroblasts cells obtained from Coriell Institute (Coriell cat#: GM9677) using Lipofectamine 2000 according to the manufacturer's instructions. Cells were plated at 1 x 10 4 cells per well of a 96 well plate. The following day the cells were transfected with their respective combination of oligo. Oligos were left on cells without media change for 48 hours prior to mRNA expression analysis.

Chromatin Immunoprecipitation (ChIP)

Cells were crosslinked with 1% formaldehyde for 10 minutes at room temperature and then quenched with glycine. Chromatin was prepared and sonicated (Covaris S200) to a size range of 300-500 bp. Antibodies for H3, H3K27me3, H3K36me3, EZH2, and RNA

Polymerase II Serine 2 (Abeam) and H3K4me3 (Millipore) were coupled to Protein G magnetic beads (NEB), washed, and then resuspended in IP blocking buffer. Chromatin lysates were added to the beads and immunoprecipitated overnight at 4°C.

Immunoprecipitations were washed, RNase A (Roche) treated, Proteinase K (Roche) treated, and the crosslinks were reversed by incubation overnight at 65°C. DNA was purified, precipitated, and resuspended in nuclease-free water. Custom Taqman probe sets were used to determine the enrichment of DNA. RESULTS:

Experiments were performed to confirm that SMN expression was regulated by PRC2 and that SMN oligos designed to target PRC2-associated regions could upregulate SMN expression. Firstly, EED, a component of the PRC2 complex, was knocked down in

GM09677 cells using 3 siRNAs specific for EED. EED siRNA were purchased from Qiagen (EED#3: catalog number SI00376299; EED#6: catalog number SI03037335). RNA was extracted from the treated cells using standard protocols. EED and SMN2 mRNA levels were measured after siRNA treatment with 50 nM concentration of siRNA for 3 days. It was found that full-length SMN2 mRNA levels were upregulated after EED siRNA treatment, confirming PRC2 involvement in SMN expression regulation (FIG. 10). Cells were then treated with SMN Oligos 77 and 83, which were designed to target PRC2- associated regions in SMN, described in Table 4 or Table 6 at 30 nM concentration for 3 days. RNA was then extracted and SMN2 levels were measured. It was found that both SMN oligos 77 and 83 upregulated SMN2 levels compared to controls (FIG. 10), confirming that oligos that target PRC2-associated regions can upregulate SMN levels.

Next, chromatin immunoprecipitation (ChIP) was used to determine the chromatin status at the SMN locus (FIG. 11A). It was found that knockdown of PRC2 components EED and EZH2 reduced the presence of EZH2 in SMN chromatin (FIG. 1 IB). Histone 3 is trimethylated at Lysine 27 (the H3K27m3 modification) by the action of EZH2, the histone methyltransferase enzyme in the PRC2 complex. This H3K27m3 modification is understood to repress transcription. EED and EZH1/2 knockdown also slightly decreased the H3K27m3 mark (FIG. 11C). The decrease in H3K27m3 was not as great as the decrease in EZH2 because it typically requires longer periods of time to observe the loss of the H3K27m3 mark. These data show that knockdown of PRC2 components leads to decreases in the presence of EZH2 and H3K27m3 in the SMN gene chromatin, indicating that PRC2 normally acts at the SMN gene. It was also found that knockdown of PRC2 components led to an increase in marks of transcriptional activity, e.g., an increase in RNA Pol II S2 and H3K36m3 within regions of the SMN locus (FIG. 12). The H3K36m3 modification is a mark of transcriptional elongation. The increase in the presence of RNA polymerase II and the H3K36m3 modification indicate that knockdown of PRC2 leads to increased SMN transcription.

Therefore, PRC2 does normally repress the transcription of the SMN gene. The HOXC13 gene was used as a positive control for the EED and EZH1/2 knockdown as it is known to be repressed by PRC2. Therefore, it was found that EED knockdown decreased EZH2, decreased the repressive H3K27 methylation mark, increased the H3K36 methylation mark of transcriptional elongation, and increased the presence of RNA polymerase II at this genomic location (FIG. 11D).

Subsequently, chromatin status was measured after treatment with SMN oligos.

GM09677 cells were treated with 30 nM concentration of SMN oligos 77 and 83 for 3 days. It was found that oligos 77 and 83 reduced EZH2 association in the SMN gene locus (FIG. 13A). The loss of EZH2 association was not observed with splice- switching oligos.

H3K27me3 was decreased with SMN oligo 83 treatment , consistent with oligo treatment blocking the recruitment of EZH2, the histone methyltransferase that applies the H3K27me3 repressive chromatin modification (FIG. 13B). Oligo treatment did not affect the HOXC 13 promoter chromatin content (FIG. 13C). This latter result indicates selectivity of the action of the oligos as they only block recruitment of PRC2 to the SMN gene, but do not affect PRC2 activity at H0XC13, another PRC2-regulated gene. SMN oligo treatment also increased marks of transcriptional elongation, e.g., H3K36m3 and RNA pol II S2, similar to the knockdown of PRC2 components (FIG. 14). An exemplary splice- switching oligo, on the other hand, did not increase marks of transcriptional elongation (FIG. 15). These results indicate that SMN upregulating oligo treatment increases transcription in a manner that is consistent with the inhibition of the recruitment of PRC2 to the SMN gene.

SMN oligos then were tested in non-human primate (NHP) cells to assess levels of SMNl expression. It was found that many SMN oligos upregulated levels of SMNl in NHP cells in a dose-dependent manner (FIG. 16A). Exon 7 exclusion is a human- specific phenomenon occurring in SMA patients, and is not observed in NHPs (Rochette CF etal. Hum Genet. 2001 Mar; 108(3):255-66. PMID: 11354640). As a result, exemplary splice switching oligos were not able to upregulate SMN expression in NHP cells (FIG. 16B).

These results confirm that oligonucleotides targeting PRC2-associated RNAs (which may be referred herein to as PRC2-targeting oligos) are capable of upregulating expression wild- type SMNl. These results are particularly relevant with respect to treatment of ALS patient through SMN upregulation, because ALS patients typically possess wild-type SMNl gene and thus would be responsive to upregulation via PRC2-targeting oligos.

Next, SMN oligos designed to target PRC2-associated regions and SMN splice- switching oligo treatments were combined to determine if having oligos with multiple mechanisms of action would have an synergistic effect. It was found that combining SMN PRC2-targeting oligos with splice- switching oligos to treat cells resulted in increased upregulation of SMN2 compared to single oligo treatment (FIG. 17).

Lastly, several SMN oligos designed to target PRC2-associated regions were tested in GM09677 cells for their ability to upregulate SMN2 mRNA and protein levels. GM09677 cells were treated with a 30 nM concentration of SMN oligos for 3 days. It was found that all oligos tested upregulated full-length SMN mRNA to some degree, with some oligos upregulating SMN mRNA over 2-fold above control levels (FIG. 18). It was also found that the oligos tested upregulated SMN protein by at least 2-fold over control levels (FIG. 19). Several splice- switching oligos were also tested in GM09677 cells for their ability to upregulate SMN2 mRNA and protein levels. GM09677 cells were treated with a 30 nM concentration of splicing oligos for 3 days. It was found that all splice- switching oligos tested upregulated full-length SMN mRNA to some degree, with some oligos upregulating SMN mRNA over 2-fold above control levels (FIG. 20). It was also found that the splice- switching oligos tested upregulated SMN protein by at least 3-fold over control levels (FIG. 21).

In summary, it was found that PRC2 knockdown increased SMN expression, RNA polymerase occupancy and activating chromatin marks (indicating transcriptional activation), and decreased EZH2 association with the SMN gene. Similarly, SMN oligos designed to target PRC2-associated regions also increased SMN1 and 2 expression, RNA polymerase occupancy and activating chromatin marks (indicating transcriptional activation), and decreased EZH2 association with a SMN gene. Further, combining oligos that involved different mechanisms of action may result in increased SMN expression compared to use of single oligos.

The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by examples provided, since the examples are intended as a single illustration of one aspect of the invention and other functionally equivalent embodiments are within the scope of the invention. Various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention.