RNA targeting of mutations via inhibitory tRNAs and deaminases

文档序号:1651973 发布日期:2019-12-24 浏览:29次 中文

阅读说明:本技术 经由抑制性tRNAs和脱氨酶对突变进行RNA靶向 (RNA targeting of mutations via inhibitory tRNAs and deaminases ) 是由 普拉尚特·马利 德拉瓦·卡特埃卡 于 2018-03-02 设计创作,主要内容包括:本公开的方面涉及一种利用修饰的tRNA的用于由终止密码子中的突变引起的疾病、病症或状况的基因治疗方法。包括肌营养不良症(例如杜兴氏肌营养不良症)、一些癌症、β型地中海贫血、赫尔勒氏综合征和囊性纤维化在内的所有遗传性疾病中的至少10%-15%落入该类别内。不受理论所束缚,认为这种方法比CRISPR方法更安全,这是因为其具有最小的脱靶效应并且缺乏基因组水平变化。(Aspects of the disclosure relate to a method of gene therapy for a disease, disorder, or condition caused by a mutation in a stop codon using a modified tRNA. At least 10% -15% of all genetic diseases including muscular dystrophy (e.g., duchenne muscular dystrophy), some cancers, beta thalassemia, heller's syndrome, and cystic fibrosis fall within this category. Without being bound by theory, this approach is believed to be safer than the CRISPR approach because it has minimal off-target effects and lacks genomic level variation.)

1.A method for restoring expression of a protein comprising a point mutation in an RNA sequence encoding the protein in a subject in need thereof, comprising administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon.

2. The method of claim 1, wherein the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA.

3. The method of claim 1 or 2, wherein the tRNA is an endogenous tRNA with a modified anticodon stem that recognizes the codon comprising the point mutation.

4. The method of claim 3, wherein the tRNA is serine-charged.

5. The method of claim 1 or 2, wherein the tRNA is an orthogonal tRNA with a non-canonical amino acid.

6. The method of claim 5, wherein the carrier further comprises a corresponding tRNA synthetase.

7. The method of claim 5 or 6, wherein the non-canonical amino acid is pyrrolysine.

8. The method of claim 7, wherein the method further comprises introducing pyrrolysine into the diet of the subject.

9. The method of any one of the preceding claims, wherein the vector encodes two trnas having anticodon sequences that recognize the codon comprising the point mutation.

10. The method of any one of the preceding claims, wherein the gene is dystrophin.

11. A method of treating a disease, disorder, or condition in a subject in need thereof, the disease, disorder, or condition characterized by the presence of a point mutation in an RNA sequence encoding a gene associated with the disease, disorder, or condition, the method comprising administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon.

12. The method of any one of claims 11, wherein the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA.

13. The method of claim 11 or 12, wherein the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes the codon comprising the point mutation.

14. The method of claim 13, wherein the tRNA carries serine.

15. The method of claim 11 or 12, wherein the tRNA is an orthogonal tRNA with a non-canonical amino acid.

16. The method of claim 15, wherein the carrier further comprises a corresponding tRNA synthetase.

17. The method of claim 15 or 16, wherein the non-canonical amino acid is pyrrolysine.

18. The method of claim 17, wherein the method further comprises introducing pyrrolysine into the diet of the subject.

19. The method of any one of claims 11-18, wherein the vector encodes two trnas having anticodon sequences that recognize codons comprising the point mutations.

20. The method of any one of claims 11 to 19, wherein the disease, disorder or condition is selected from the group consisting of the diseases, disorders and conditions listed in table 1, optionally characterized by the presence of a nonsense mutation and/or a premature stop codon.

21. The method of any one of claims 11-20, wherein the gene is dystrophin.

22. The method of claim 21, wherein the disease, disorder, or condition is muscular dystrophy.

23. The method of claim 22, wherein the muscular dystrophy is duchenne muscular dystrophy.

24. A vector encoding one or more trnas having an anticodon sequence that recognizes a codon in an RNA sequence encoding a gene that comprises a point mutation, optionally wherein the point mutation produces a premature stop codon.

25. The vector of claim 24, wherein the point mutation generates a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA.

26. The vector of claim 24 or 25, wherein the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes the codon comprising the point mutation.

27. The vector of claim 26, wherein the tRNA carries serine.

28. The vector of claim 24 or 25, wherein the tRNA is an orthogonal tRNA with a non-canonical amino acid.

29. The vector of claim 28, wherein the vector further comprises a corresponding tRNA synthetase.

30. The vector according to claim 28 or 29, wherein the non-canonical amino acid is pyrrolysine.

31. The vector according to any one of claims 24 to 30, wherein the vector is an AAV vector.

32. The vector of claim 31, wherein the vector is an AAV8 vector.

33. The vector of any one of claims 24-32, wherein the vector encodes two trnas having anticodon sequences that recognize the codon comprising the point mutation.

34. The vector of any one of claims 24-33, wherein the gene is dystrophin.

35. A method for restoring expression of a protein comprising a point mutation in an RNA sequence encoding the protein in a subject in need thereof, the method comprising administering to the subject one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for the ADAR and one or more corresponding reverse guide RNAs ("radrnas") for the ADAR, wherein the ADAR-based RNA editing system specifically edits the point mutation.

36. The method of claim 35, wherein the point mutation generates a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA.

37. The method of claim 36, wherein the ADAR-based RNA editing system converts UAA to UIA.

38. The method of claim 37, wherein the ADAR-based RNA editing system converts UIA to UII.

39. The method of claim 36, wherein the ADAR-based RNA editing system converts UAA to UAI.

40. The method of claim 39, wherein the one or more vectors further encode a tRNA that targets amber codons.

41. The method of any one of claims 35-40, wherein the protein is dystrophin.

42. The method of claim 35, wherein the point mutation generates a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG.

43. The method of claim 42, wherein the ADAR-based RNA editing system converts CAG to CIG.

44. The method of any one of claims 35, 42, or 43, wherein the protein is ornithine transcarbamylase.

45. The method of any one of claims 35 to 44, wherein the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E1008Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and an inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC.

46. The method of claim 45, wherein the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide.

47. The method of any one of claims 35 to 46, wherein the method further comprises administering an effective amount of interferon to enhance endogenous ADAR1 expression.

48. The method of claim 47, wherein the interferon is interferon alpha.

49. The method of any one of claims 35-48, wherein the adrA comprises one or more RNA hairpin motifs.

50. The method of claim 49, wherein the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops.

51. The method of claim 49, wherein the one or more RNA hairpin motifs are stabilized by replacing A-U with G-C.

52. The method of any one of claims 35-51, wherein the adRNA is stabilized via incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

53. A method of treating a disease, disorder, or condition characterized by the presence of a point mutation in an RNA sequence encoding a protein associated with the disease, disorder, or condition in a subject in need thereof, comprising administering to the subject one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for the ADAR and one or more corresponding reverse guide RNAs ("radrnas") for the ADAR, wherein the ADAR-based RNA editing system specifically edits the point mutation.

54. The method of claim 53, wherein the point mutation generates a nonsense mutation having the DNA sequence TAA and the RNA sequence UAA.

55. The method of claim 54, wherein the ADAR-based RNA editing system converts UAA to UIA.

56. The method of claim 53, wherein the ADAR-based RNA editing system converts UIA to UII.

57. The method of claim 54, wherein the ADAR-based RNA editing system converts UAA to UAI.

58. The method of claim 57, wherein the one or more vectors further encode a tRNA that targets amber codons.

59. The method of any one of claims 53-58, wherein the protein is dystrophin.

60. The method of claim 53, wherein the point mutation generates a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG.

61. The method of claim 60, wherein the ADAR-based RNA editing system converts CAG to CIG.

62. The method of any one of claims 53, 60, or 61, wherein said protein is ornithine transcarbamylase.

63. The method of any one of claims 53-62, wherein the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E1008Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and an inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC.

64. The method of claim 63, wherein the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide.

65. The method of any one of claims 53 to 64, wherein the method further comprises administering an effective amount of interferon to enhance endogenous ADAR1 expression.

66. The method of claim 65, wherein the interferon is interferon alpha.

67. The method of any one of claims 53-66, wherein the adrA comprises one or more RNA hairpin motifs.

68. The method of claim 67, wherein the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops.

69. The method of claim 68, wherein the one or more RNA hairpin motifs are stabilized by replacing A-U with G-C.

70. The method of any one of claims 50-69, wherein the adRNA is stabilized via incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

71. The method of any one of claims 53 to 70, wherein the disease, disorder or condition is selected from the group consisting of the diseases, disorders and conditions listed in Table 1.

72. The method of claim 71, wherein the protein is dystrophin and the disease, disorder, or condition is muscular dystrophy.

73. The method of claim 72, wherein the muscular dystrophy is Duchenne muscular dystrophy.

74. A recombinant expression system comprising one or more vectors encoding an ADAR-based RNA editing system for a subject, the ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for the ADAR and one or more corresponding reverse guide RNAs ("radrnas") for the ADAR, wherein the ADAR-based RNA editing system specifically edits a point mutation in an RNA sequence encoding a protein.

75. The recombinant expression system of claim 74, wherein the point mutation generates a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA.

76. The recombinant expression system of claim 75, wherein the ADAR-based RNA editing system converts UAA to UIA.

77. The recombinant expression system of claim 76, wherein the ADAR 2-based RNA editing system converts UIA to UII.

78. The recombinant expression system of claim 75, wherein the ADAR 2-based RNA editing system converts UAA to UAI.

79. The recombinant expression system of claim 78, wherein the one or more vectors further encode a tRNA that targets amber codons.

80. The recombinant expression system of any one of claims 74-79, wherein the protein is dystrophin.

81. The recombinant expression system of claim 74, wherein said point mutation generates a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG.

82. The recombinant expression system of claim 81, wherein the ADAR-based RNA editing system converts CAG to CIG.

83. The recombinant expression system of any one of claims 74, 81, or 82, wherein said protein is ornithine transcarbamylase.

84. The recombinant expression system of any one of claims 74-83, wherein the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E1008Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC.

85. The recombinant expression system of claim 84, wherein the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide.

86. The recombinant expression system of any one of claims 74-85, wherein the adRNA comprises one or more RNA hairpin motifs.

87. The recombinant expression system of claim 86, wherein the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops.

88. The recombinant expression system of claim 86, wherein the one or more RNA hairpin motifs are stabilized by replacing A-U with G-C.

89. The recombinant expression system of any one of claims 74-88, wherein the adRNA is stabilized via incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

90. The recombinant expression system of any one of claims 74-89, wherein the protein is dystrophin or ornithine transcarbamylase.

91. A composition comprising the recombinant expression system of any one of claims 74-90 and an effective amount of interferon to enhance expression of endogenous ADAR 1.

92. The composition of claim 91, wherein the interferon is interferon alpha.

Background

Aspects of the disclosure relate to a method of gene therapy for a disease, disorder, or condition caused by a mutation in a stop codon using a modified tRNA. At least 10% -15% of all genetic diseases including muscular dystrophy (e.g., duchenne muscular dystrophy), some cancers, beta-thalassemia, Hurler's syndrome (Hurler syndrome), and cystic fibrosis fall within this category. Without being bound by theory, this approach is believed to be safer than the CRISPR method or TALEN method because it has minimal off-target effects and lacks genomic level changes.

Disclosure of Invention

Aspects of the present disclosure relate to a method for restoring expression of a protein comprising a point mutation in an RNA sequence encoding the protein in a subject in need thereof, the method comprising, or alternatively consisting essentially of, or yet further consisting of: administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In additional embodiments, pyrrolysine is administered to the subject by introduction into the subject's diet. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the protein is dystrophin. In another aspect, the subject is a human and optionally a pediatric patient.

A further method aspect relates to treating a disease, disorder or condition in a subject in need thereof characterized by the presence of a point mutation in an RNA sequence encoding a protein associated with the disease, disorder or condition, the method comprising, or alternatively consisting essentially of, or yet further consisting of: administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In further embodiments, pyrrolysine is introduced into the subject's diet. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the disease, disorder or condition is selected from the group consisting of the diseases, disorders and conditions listed in table 1, optionally characterized by the presence of a nonsense mutation and/or a premature stop codon. In some embodiments, the protein is dystrophin. In additional embodiments, the disease, disorder, or condition is muscular dystrophy. In yet further embodiments, the disease, disorder, or condition is duchenne muscular dystrophy. In some embodiments, the subject is a human and optionally a pediatric patient.

Still further aspects disclosed herein relate to a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising a point mutation in an RNA sequence encoding a protein, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the vector is an AAV vector, optionally an AAV8 vector. In some embodiments, the protein is dystrophin. In another aspect, the subject is a human and optionally a pediatric patient.

In another aspect, the present disclosure relates to a method for restoring protein expression in a subject in need thereof, said protein comprising a point mutation in an RNA sequence encoding said protein, said method comprising administering to said subject one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adras") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR, wherein said ADAR-based RNA editing system specifically edits said point mutation. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, optionally in embodiments involving nonsense or missense mutations, the targeted RNA is mRNA. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the protein is dystrophin. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In some embodiments, optionally in embodiments involving splice site mutations, the targeted RNA is a precursor mRNA. In some embodiments, the protein is ornithine transcarbamylase. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the method further comprises administering an effective amount of interferon to enhance endogenous ADAR1 expression. In yet further embodiments, the interferon is interferon alpha. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA. In another aspect, the subject is a human and optionally a pediatric patient.

A further method aspect relates to a method of treating a disease, disorder or condition in a subject in need thereof, characterized by the presence of a point mutation in an RNA sequence encoding a protein associated with the disease, disorder or condition, the method comprising, or alternatively consisting essentially of, or yet further consisting of: administering to the subject one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adRNAs") for ADAR and one or more corresponding reverse guide RNAs ("radRNAs") for ADAR, wherein the ADAR-based RNA editing system specifically edits the point mutation. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, optionally in embodiments involving nonsense or missense mutations, the targeted RNA is mRNA. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the protein is dystrophin. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In some embodiments, optionally in embodiments involving splice site mutations, the targeted RNA is a precursor mRNA. In some embodiments, the protein is ornithine transcarbamylase. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the method further comprises administering an effective amount of interferon to enhance endogenous ADAR1 expression. In yet further embodiments, the interferon is interferon alpha. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA. In some embodiments, the disease, disorder, or condition is selected from the group consisting of the diseases, disorders, and conditions listed in table 1. In additional embodiments, the protein is dystrophin and the disease, disorder, or condition is muscular dystrophy. In yet further embodiments, the disease, disorder, or condition is duchenne muscular dystrophy. In some embodiments, the subject is a human and optionally a pediatric patient.

A further aspect relates to a recombinant expression system comprising one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR for a subject, wherein the ADAR-based RNA editing system specifically edits a point mutation in an RNA sequence encoding a protein. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, optionally in embodiments involving nonsense or missense mutations, the targeted RNA is mRNA. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the protein is dystrophin. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In some embodiments, optionally in embodiments involving splice site mutations, the targeted RNA is a precursor mRNA. In some embodiments, the protein is ornithine transcarbamylase. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA. In another aspect, the subject is a human and optionally a pediatric patient.

Yet a further aspect relates to a composition comprising any one or more of the vectors disclosed herein and optionally one or more carriers, such as a pharmaceutically acceptable carrier. In some embodiments, the composition further comprises an effective amount of interferon to enhance endogenous ADAR1 expression. In yet further embodiments, the interferon is interferon alpha.

Some aspects disclosed herein relate to a method for restoring protein expression in a subject in need thereof, the method comprising, or alternatively consisting essentially of, or yet further consisting of: administering to the subject a tRNA having an anticodon sequence that recognizes a mutation in an RNA sequence encoding the protein or administering to the subject a vector encoding one or more of the trnas. In some embodiments, the mutation is a nonsense mutation, optionally a premature stop codon. In some embodiments, the nonsense mutation is a TAA in DNA and a UAA in RNA. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, a non-canonical amino acid is introduced or administered to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein is dystrophin. In another aspect, the subject is a human and optionally a pediatric patient.

Further aspects disclosed herein relate to a method of treating a disease, disorder or condition characterized by protein deficiency in a subject in need thereof, the method comprising, or alternatively consisting essentially of, or yet further consisting of: administering to the subject a tRNA having an anticodon sequence that recognizes a mutation in an RNA sequence that encodes the protein or a vector that encodes one or more of the tRNA. In some embodiments, the mutation is a nonsense mutation, optionally a premature stop codon. In some embodiments, the nonsense mutation is a TAA in DNA and a UAA in RNA. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, a non-canonical amino acid is administered or introduced to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein deficiency is a dystrophin deficiency. In some embodiments, the disease, disorder, or condition is muscular dystrophy. In some embodiments, the muscular dystrophy is duchenne muscular dystrophy. In another aspect, the subject is a human and optionally a pediatric patient.

Other aspects relate to a vector encoding one or more trnas having an anticodon sequence that recognizes a mutation in an RNA sequence encoding the protein. In some embodiments, the mutation is a nonsense mutation, optionally a premature stop codon. In some embodiments, the nonsense mutation is a TAA in DNA and a UAA in RNA. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, the vector further comprises a corresponding synthetase. In some embodiments, a non-canonical amino acid is introduced or administered to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein is dystrophin. In some embodiments, the mutation is a nonsense mutation, optionally a premature stop codon. In some embodiments, the vector is an adeno-associated virus (AAV) vector. In some embodiments, the AAV vector is an AAV8 vector.

Additional aspects of the present disclosure relate to on-demand in vivo production of therapeutic proteins, such as, but not limited to, (i) insulin; (ii) neutralizing antibodies against viruses (e.g., HIV, HCV, HPV, influenza) and bacteria (e.g., Staphylococcus aureus; drug resistant strains). These method aspects include administering to a subject a vector encoding a therapeutic protein having a mutation in the sequence and a tRNA having an anticodon sequence that recognizes the mutation in an RNA sequence encoding the therapeutic protein or a vector encoding one or more of the trnas. Thus, any of the methods and vectors disclosed above relating to trnas having anticodon sequences that recognize mutations in RNA sequences encoding proteins, or vectors encoding one or more of said trnas, can also be applied to this aspect.

Some aspects disclosed herein relate to a method for restoring protein expression in a subject in need thereof, the method comprising administering to the subject an ADAR 2-based RNA editing system comprising ADAR2, one or more forward guide RNAs ("adrnas") for ADAR2, and one or more corresponding reverse guide RNAs ("radrnas") for ADAR2, wherein the ADAR 2-based RNA editing system specifically edits mutations in an RNA sequence encoding the protein; or one or more vectors encoding the ADAR2, an adRNA, a radRNA. In some embodiments, the ADAR 2-based RNA editing system changes adenosine (a) to inosine (I), which is read as guanosine (G) during translation. In some embodiments, the mutation is a nonsense mutation. In some embodiments, the nonsense mutation is a TAA in DNA and a UAA in RNA. In some embodiments, the ADAR 2-based RNA editing system causes a point mutation at one or more adenosines (a) in a nonsense mutation. In some embodiments, the ADAR 2-based RNA editing system converts UAA to UIA (read as UGA). In further embodiments, the ADAR 2-based RNA editing system converts the UIA (read as UGA) to the UII (read as UGG). In some embodiments, the ADAR 2-based RNA editing system converts UAA to UAI (read as UAG). In some embodiments, the method further comprises administering a tRNA, such as a tRNA disclosed above, that recognizes a codon encoded by a sequence edited by ADAR 2. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, a non-canonical amino acid is introduced to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein deficiency is a dystrophin deficiency. In some embodiments, the disease, disorder, or condition is muscular dystrophy. In some embodiments, the muscular dystrophy is duchenne muscular dystrophy.

A further aspect disclosed herein relates to a method of treating a disease, disorder or condition characterized by protein deficiency in a subject in need thereof, the method comprising administering to the subject an ADAR 2-based RNA editing system comprising ADAR2, one or more forward guide RNAs ("adRNA") for ADAR2, and one or more corresponding reverse guide RNAs ("radRNA") for ADAR2, wherein the ADAR 2-based RNA editing system specifically edits a mutation in an RNA sequence encoding the protein or one or more vectors encoding the ADAR2, adRNA, radRNA. In some embodiments, the ADAR 2-based RNA editing system changes adenosine (a) to inosine (I), which is read as guanosine (G) during translation. In some embodiments, the mutation is a nonsense mutation. In some embodiments, the nonsense mutation is a TAA. In some embodiments, the ADAR 2-based RNA editing system causes a point mutation at one or more adenosines (a) in a nonsense mutation. In some embodiments, the ADAR 2-based RNA editing system converts UAA to UIA (read as UGA). In further embodiments, the ADAR 2-based RNA editing system converts the UIA (read as UGA) to the UII (read as UGG). In some embodiments, the ADAR 2-based RNA editing system converts UAA to UAI (read as UAG). In some embodiments, the method further comprises administering a tRNA, such as a tRNA disclosed above, that recognizes a codon encoded by a sequence edited by ADAR 2. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, a non-canonical amino acid is introduced to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein deficiency is a dystrophin deficiency. In some embodiments, the disease, disorder, or condition is muscular dystrophy. In some embodiments, the muscular dystrophy is duchenne muscular dystrophy.

Other aspects relate to a recombinant expression system comprising one or more vectors encoding an ADAR 2-based RNA editing system comprising one or more of one or more ADAR2, one or more forward guide RNAs ("adrnas") for ADAR2, and one or more corresponding reverse guide RNAs ("radrnas") for ADAR2, wherein the ADAR 2-based RNA editing system specifically edits mutations in an RNA sequence encoding a protein. In some embodiments, ADAR2 changes adenosine (a) to inosine (I), which is read during translation to guanosine (G). In some embodiments, an adRNA/radRNA pair directs the transformation of UAA into UIA (read as UGA). In a further embodiment, the second adRNA/radRNA pair directs the transformation of the UIA (read as UGA) to the UII (read as UGG). In some embodiments, an adRNA/radRNA pair directs the transformation of UAA to UAI (read as UAG). In some embodiments, one or more vectors or additional vectors further encode a tRNA, such as a tRNA disclosed above, that recognizes a codon encoded by a sequence edited by ADAR 2. In some embodiments, the tRNA is a modified endogenous tRNA with a canonical amino acid. In some embodiments, the canonical amino acid is serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In some embodiments, the orthogonal tRNA has a corresponding synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments, a non-canonical amino acid is introduced to the subject (e.g., via food), thereby allowing orthogonal tRNA activity to be induced. In some embodiments, the non-canonical amino acid is pyrrolysine. In some embodiments, the tRNA targets an amber codon. In some embodiments, the tRNA targets an ochre-type codon. In some embodiments, the tRNA targets an opal-type codon. In some embodiments, the protein is dystrophin. In some embodiments, the mutation is a nonsense mutation. In some embodiments, the vector is an adeno-associated virus (AAV) vector. In some embodiments, the AAV vector is an AAV8 vector.

Additional aspects of the present disclosure relate to on-demand in vivo production of therapeutic proteins, such as, but not limited to, (i) insulin; (ii) neutralizing antibodies against viruses (e.g., HIV, HCV, HPV, influenza) and bacteria (e.g., Staphylococcus aureus; drug resistant strains). These method aspects include administering to a subject a vector encoding a therapeutic protein having a mutation in the sequence and an ADAR 2-based RNA editing system comprising ADAR2, one or more forward guide RNAs ("adrnas") for ADAR2, and one or more corresponding reverse guide RNAs ("radrnas") for ADAR2, wherein the ADAR 2-based RNA editing system specifically edits the mutation in the RNA sequence encoding the protein or edits the one or more vectors encoding the ADAR2, adrnas, radrnas. Thus, any of the methods and vectors disclosed above relating to ADAR 2-based RNA editing systems specifically edit mutations in the RNA sequence encoding the protein, or edit one or more vectors encoding the ADAR2, adRNA, radRNA.

Partial sequence listing

mU6, tRNA (U25C) amber

tcccggggtttccgccaTTTTTTGGTACTGAGtCGCCCaGTCTCAGATAGATCCGACGCCGCCATCTCTAGGCCCGCGCCGGCCCCCTCGCACAGACTTGTGGGAGAAGCTCGGCTACTCCCCTGCCCCGGTTAATTTGCATATAATATTTCCTAGTAACTATAGAGGCTTAATGTGCGATAAAAGACAGATAATCTGTTCTTTTTAATACTAGCTACATTTTACATGATAGGCTTGGATTTCTATAAGAGATACAAATACTAAATTATTATTTTAAAAAACAGCACAAAAGGAAACTCACCCTAACTGTAAAGTAATTGTGTGTTTTGAGACTATAAATATCCCTTGGAGAAAAGCCTTGTTTGggaaacctgatcatgtagatcgaaCggactCTAaatccgttcagccgggttagattcccggggtttccgccaTTTTTTCCTAGACCCAGCTTTCTTGTACAAAGTTGG

mU6, tRNA (U25C) ochre form

tcccggggtttccgccaTTTTTTGGTACTGAGtCGCCCaGTCTCAGATAGATCCGACGCCGCCATCTCTAGGCCCGCGCCGGCCCCCTCGCACAGACTTGTGGGAGAAGCTCGGCTACTCCCCTGCCCCGGTTAATTTGCATATAATATTTCCTAGTAACTATAGAGGCTTAATGTGCGATAAAAGACAGATAATCTGTTCTTTTTAATACTAGCTACATTTTACATGATAGGCTTGGATTTCTATAAGAGATACAAATACTAAATTATTATTTTAAAAAACAGCACAAAAGGAAACTCACCCTAACTGTAAAGTAATTGTGTGTTTTGAGACTATAAATATCCCTTGGAGAAAAGCCTTGTTTGggaaacctgatcatgtagatcgaaCggactTTAaatccgttcagccgggttagattcccggggtttccgccaTTTTTTCCTAGACCCAGCTTTCTTGTACAAAGTTGG

mU6, tRNA (U25C) opal form

tcccggggtttccgccaTTTTTTGGTACTGAGtCGCCCaGTCTCAGATAGATCCGACGCCGCCATCTCTAGGCCCGCGCCGGCCCCCTCGCACAGACTTGTGGGAGAAGCTCGGCTACTCCCCTGCCCCGGTTAATTTGCATATAATATTTCCTAGTAACTATAGAGGCTTAATGTGCGATAAAAGACAGATAATCTGTTCTTTTTAATACTAGCTACATTTTACATGATAGGCTTGGATTTCTATAAGAGATACAAATACTAAATTATTATTTTAAAAAACAGCACAAAAGGAAACTCACCCTAACTGTAAAGTAATTGTGTGTTTTGAGACTATAAATATCCCTTGGAGAAAAGCCTTGTTTGggaaacctgatcatgtagatcgaaCggactTCAaatccgttcagccgggttagattcccggggtttccgccaTTTTTTCCTAGACCCAGCTTTCTTGTACAAAGTTGG

MmPylRS(AfIII)

CAGCCTCCGGACTCTAGAGGATCGAACCCTTAAGgccaccATGGATAAGAAACCTTTGAACACTCTCATTAGTGCGACAGGGCTCTGGATGTCCCGAACGGGGACTATACACAAGATAAAACACCATGAGGTCTCAAGGAGCAAAATCTATATCGAGATGGCATGCGGCGACCATCTTGTGGTAAATAATAGTAGGTCCTCCAGGACGGCAAGAGCACTCCGACATCACAAGTACAGAAAAACCTGCAAACGGTGTAGGGTATCCGACGAAGACTTGAACAAATTTTTGACTAAGGCCAACGAGGATCAAACTTCTGTCAAAGTGAAAGTGGTTTCTGCTCCTACCCGAACTAAGAAGGCCATGCCCAAGTCCGTGGCAAGGGCACCCAAGCCACTCGAAAATACTGAGGCCGCTCAGGCCCAACCATCCGGTAGTAAGTTCAGTCCAGCCATACCCGTAAGTACCCAAGAATCTGTCAGTGTGCCGGCCTCAGTTTCCACATCTATAAGTTCAATTTCTACAGGAGCGACGGCCTCCGCCCTCGTCAAGGGTAACACAAACCCGATAACTTCTATGAGTGCCCCCGTACAGGCATCCGCACCAGCACTGACGAAGTCTCAAACTGATAGGCTGGAAGTGCTCTTGAATCCGAAGGACGAGATATCTCTTAACTCCGGTAAACCTTTCCGGGAGCTGGAAAGTGAACTTCTCAGCCGGCGAAAAAAAGACCTCCAGCAAATTTACGCAGAGGAAAGGGAGAACTATCTGGGGAAGTTGGAACGAGAGATCACCCGATTCTTTGTCGATCGCGGATTTTTGGAGATTAAAAGCCCAATTCTCATCCCCCTTGAATATATCGAACGAATGGGAATCGACAATGATACGGAGTTGTCCAAGCAGATTTTCCGCGTAGACAAGAACTTTTGTCTTCGACCCATGCTCGCTCCGAACCTCTACAATTACTTGAGAAAGTTGGACAGAGCGCTCCCGGACCCGATCAAGATATTTGAGATCGGTCCTTGTTATAGAAAGGAGAGTGATGGAAAAGAACACCTCGAAGAGTTCACGATGCTGAACTTCTGCCAAATGGGTTCTGGCTGCACACGGGAGAATCTCGAAAGCATCATTACAGATTTCCTTAACCATCTGGGGATAGACTTTAAAATAGTGGGTGACAGCTGTATGGTATACGGAGATACCTTGGACGTAATGCACGGGGATCTTGAGCTTTCCTCCGCCGTGGTTGGACCTATACCGTTGGACCGGGAGTGGGGAATCGACAAACCGTGGATAGGCGCCGGTTTCGGCCTTGAAAGACTCCTCAAAGTCAAGCATGATTTCAAAAACATAAAACGGGCTGCTCGCTCCGAATCTTATTACAACGGTATAAGTACGAACCTGTGATAATAGCTTAAGGGTTCGATCCCTACtGGTTAGTAATGAGTTTA

tRNAs

Amber inhibition:

ggaaacctgatcatgtagatcgaatggactctaaatccgttcagccgggttagattcccggggtttccgcca

amber inhibition (2):

ggggggtggatcgaatagatcacacggactctaaattcgtgcaggcgggtgaaactcccgtactccccgcca

ochre type inhibition:

ggaaacctgatcatgtagatcgaatggactttaaatccgttcagccgggttagattcccggggtttccgcca opal-type inhibition:

ggaaacctgatcatgtagatcgaatggacttcaaatccgttcagccgggttagattcccggggtttccgcca

synthesizing enzyme:

ATGGATAAAAAACCATTAGATGTTTTAATATCTGCGACCGGGCTCTGGATGTCCAGGACTGGCACGCTCCACAAAATCAAGCACCATGAGGTCTCAAGAAGTAAAATATACATTGAAATGGCGTGTGGAGACCATCTTGTTGTGAATAATTCCAGGAGTTGTAGAACAGCCAGAGCATTCAGACATCATAAGTACAGAAAAACCTGCAAACGATGTAGGGTTTCGGACGAGGATATCAATAATTTTCTCACAAGATCAACCGAAAGCAAAAACAGTGTGAAAGTTAGGGTAGTTTCTGCTCCAAAGGTCAAAAAAGCTATGCCGAAATCAGTTTCAAGGGCTCCGAAGCCTCTGGAAAATTCTGTTTCTGCAAAGGCATCAACGAACACATCCAGATCTGTACCTTCGCCTGCAAAATCAACTCCAAATTCGTCTGTTCCCGCATCGGCTCCTGCTCCTTCACTTACAAGAAGCCAGCTTGATAGGGTTGAGGCTCTCTTAAGTCCAGAGGATAAAATTTCTCTAAATATGGCAAAGCCTTTCAGGGAACTTGAGCCTGAACTTGTGACAAGAAGAAAAAACGATTTTCAGCGGCTCTATACCAATGATAGAGAAGACTACCTCGGTAAACTCGAACGTGATATTACGAAATTTTTCGTAGACCGGGGTTTTCTGGAGATAAAGTCTCCTATCCTTATTCCGGCGGAATACGTGGAGAGAATGGGTATTAATAATGATACTGAACTTTCAAAACAGATCTTCCGGGTGGATAAAAATCTCTGCTTGAGGCCAATGCTTGCCCCGACTCTTTACAACTATCTGCGAAAACTCGATAGGATTTTACCAGGCCCAATAAAAATTTTCGAAGTCGGACCTTGTTACCGGAAAGAGTCTGACGGCAAAGAGCACCTGGAAGAATTTACTATGGTGAACTTCTGTCAGATGGGTTCGGGATGTACTCGGGAAAATCTTGAAGCTCTCATCAAAGAGTTTCTGGACTATCTGGAAATCGACTTCGAAATCGTAGGAGATTCCTGTATGGTCTTTGGGGATACTCTTGATATAATGCACGGGGACCTGGAGCTTTCTTCGGCAGTCGTCGGGCCAGTTTCTCTTGATAGAGAATGGGGTATTGACAAACCATGGATAGGTGCAGGTTTTGGTCTTGAACGCTTGCTCAAGGTTATGCACGGCTTTAAAAACATTAAGAGGGCATCAAGGTCCGAATCTTACTATAATGGGATTTCAACCAATCTGTAA

EGFP:

EGFP amber type:

EGFP ochre type:

EGFP opal type:

MbPylRS

MmPylRS(uniprot)

PylT (amber type)

ggaaacctgatcatgtagatcgaaCggactCTAaatccgttcagccgggttagattcccggggtttccgccaTTTTTT

PylT (Haematitum type)

ggaaacctgatcatgtagatcgaaCggactTTAaatccgttcagccgggttagattcccggggtttccgccaTTTTTT

PylT (opal type)

ggaaacctgatcatgtagatcgaaCggactTCAaatccgttcagccgggttagattcccggggtttccgccaTTTTTT

Mouse U6 primer

tcccggggtttccgccaTTTTTTGGTACTGAGtCGCCCaGTCTCAGAT

CAAACAAGGCTTTTCTCCAAGGGATAT

tRNA (U25C) amber _ F:

general reversal:

PylT

ggaaacctgatcatgtagatcgaatggactCTAaatccgttcagccgggttagattcccggggtttccgcca

PylT*(U25C)

ggaaacctgatcatgtagatcgaaCggactCTAaatccgttcagccgggttagattcccggggtttccg cca

arg tRNA (opal type) (E-cadherin paper)

Arg tRNA (opal type) (xeroderma paper)

3. Serine tRNA (amber type)

4. Leucine tRNA (amber type)

Forward direction:

and (3) reversing:

tRNA Leu AmF (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Leu Oc F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Leu Op F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Leu R (overlap with vector, bold; anticodon sequence,bold underline):

tRNA _ Ser _ Am _ F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA _ Ser _ Oc _ F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Ser Op (overlap with vector, bold; anticodon sequence,bold underline):

tRNA _ Ser _ R (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Arg-Am-F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Arg Oc F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Arg Op F (overlap with vector, bold; anticodon sequence,bold underline):

tRNA Arg R (overlap with vector, bold; anticodon sequence,bold underline):

mU6_tRNA_ser_oc:GTACTGAGtCGCCCaGTCTCAGATAGATCCGACGCCGCCATCTCTAGGCCCGCGCCGGCCCCCTCGCACAGACTTGTGGGAGAAGCTCGGCTACTCCCCTGCCCCGGTTAATTTGCATATAATATTTCCTAGTAACTATAGAGGCTTAATGTGCGATAAAAGACAGATAATCTGTTCTTTTTAATACTAGCTACATTTTACATGATAGGCTTGGATTTCTATAAGAGATACAAATACTAAATTATTATTTTAAAAAACAGCACAAAAGGAAACTCACCCTAACTGTAAAGTAATTGTGTGTTTTGAGACTATAAATATCCCTTGGAGAAAAGCCTTGTTTGGTAGTCGTGGCCGAGTGGTTAAGGCGATGGACTTTAAATCCATTGGGGTTTCCCCGCGCAGGTTCGAATCCTGCCGACTACGTTTTTT

mU6_ tRNA _ ser _ oc _ Nhe1_ insert _ F:

AATCCTGCCGACTACGTTTTTTGTACTGAGtCGCCCAGTCT

an adra (early stop codon target, bold; edited bases,bold underline):

And (4) sequential editing:

double editing:

radRNA (premature stop codon target, bold; edited bases,bold underline):

And (4) sequential editing:

double editing:

OTC target (base edited, bold):

optimization of length of adRNA and distance of editing zone from ADAR2 recruitment domain (length of adRNA-distance of editing zone from ADAR2 recruitment domain):

16-5:atgccaccTGGggcaa

16-6:tgccaccTGGggcaag

16-7:gccaccTGGggcaagc

18-6:gatgccaccTGGggcaag

20-6:gcgatgccaccTGGggcaag

ADAR2 recruitment zone v 1:

GGGTGGAATAGTATAACAATATGCTAAATGTTGTTATAGTATCCCACCT

ADAR2 recruitment zone v 2:

GTGGAATAGTATAACAATATGCTAAATGTTGTTATAGTATCCCAC

hairpin (3') (fig. 8): GGGCCCTCTTCAGGGCCCTCTAGA

Hairpin (3') (fig. 10): atcgccctgaaaag

Fulcrum (5'): gccaccTGGgg

List of inhibitory tRNA sequences:

NNN: anticodon

In the endogenous tRNA, byNNNThe positions include complementary sequences that modify the tRNA to recognize a codon that includes a point mutation. As set forth in more detail below, in amber, ochre and opal tRNAsNNNThe sequence is as follows: amber type:NNNCTA; ochre type:NNNTCA; opal type:NNN=TTA。

primer list for Next Generation Sequencing (NGS) analysis.

Name (R) Sequences (5 'to 3')
NGS_DMD_F1 GTGTTACTGAATATGAAATAATGGAGGA
NGS_DMD_R1 ATTTCTGGCATATTTCTGAAGGTG
NGS_DMD_F2 CTCTCTGTACCTTATCTTAGTGTTACTGA
NGS_DMD_R2 CTCTTCAAATTCTGACAGATATTTCTGGC
NGS_OTC_F ACCCTTCCTTTCTTACCACACA
NGS_OTC_R CAGGGTGTCCAGATCTGATTGTT
NGS_OTC_R2 CTTCTCTTTTAAACTAACCCATCAGAGTT

List of adra antisense sequences and corresponding ADAR2 recruited scaffolds for in vivo RNA editing studies. In some embodiments, the recruitment scaffold v2 disclosed in paragraph [0083] is used with these sequences.

Name (R) AdRNA antisense sequence (3 'to 5')
OTC TGTCTGTGGCGAGCCAAACA
DMD ACTTTCTCGTTACCTTACCG

MCP-linker-ADAR1NLS (optional sequences in parentheses)

MCP-linker-ADAR2(optional sequences in parentheses)

N22 p-linker-ADAR1NLS (optional sequences in parentheses)

Nuclear localization sequence-linker-N22 p-linker-ADAR2(optional sequences in parentheses)

MCP-linker-ADAR1(E1008Q)NLS (optional sequences in parentheses)

Nuclear localization sequence-linker-MCP-linker-ADAR2(E488Q)(optional sequences in parentheses)

N22 p-linker-ADAR1(E1008Q)(optional sequences in parentheses)

Nuclear localization sequence-linker-N22 p-linker-ADAR 2(E488Q)

Nuclear localization sequence-linker-MCP-linker-hAPOPEC1

Nuclear localization sequence-linker-MCP-linker-rAPOBEC1

dsRBD-linker-rAPOBEC1

dsRBD-linker-hAPOBEC1

MCP-linker-ADAR1-NES

MCP-linker-ADAR2-NLS

MCP-linker-ADAR2-NES

MCP-linker-rAPOBEC1-NLS

MCP-linker-rAPOBEC1-NES

MCP-linker-hAPOBEC1-NLS

MCP-linker-hAPOBEC1-NES

Alternative spacers (which may be used instead of GGSGGSGGS):

SGSETPGTSESATPES

N22p-hAPOBEC1

3XNLS-4xlN-hAPOBEC1

c-terminal ADAR2 (residues 1-138 deleted)

MLRSFVQFPNASEAHLAMGRTLSVNTDFTSDQADFPDTLFNGFETPDKAEPPFYVGSNGDDSFSSSGDLSLSASPVPASLAQPPLPVLPPFPPPSGKNPVMILNELRPGLKYDFLSESGESHAKSFVMSVVVDGQFFEGSGRNKKLAKARAAQSALAAIFNLHLDQTPSRQPIPSEGLQLHLPQVLADAVSRLVLGKFGDLTDNFSSPHARRKVLAGVVMTTGTDVKDAKVISVSTGTKCINGEYMSDRGLALNDCHAEIISRRSLLRFLYTQLELYLNNKDDQKRSIFQKSERGGFRLKENVQFHLYISTSPCGDARIFSPHEPILEEPADRHPNRKARGQLRTKIESGEGTIPVRSNASIQTWDGVLQGERLLTMSCSDKIARWNVVGIQGSLLSIFVEPIYFSSIILGSLYHGDHLSRAMYQRISNIEDLPPLYTLNKPLLSGISNAEARQPGKAPNFSVNWTVGDSAIEVINATTGKDELGRASRLCKHALYCRWMRVHGKVPSHLLRSKITKPNVYHESKLAAKEYQAAKARLFTAFIKAGLGAWVEKPTEQDQFSLTP*

MS2-RNA:

Singly:

NNNNNNNNNNNNNNNNNNNNggccAACATGAGGATCACCCATGTCTGCAGggcc

double:

aACATGAGGATCACCCATGTcNNNNNNNNNNNNNNNNNNNNaACATGAGGATCACCCATGTc

BoxB RNA:

singly:

NNNNNNNNNNNNNNNNNNNNgggccctgaagaagggccc

double:

ggGCCCTGAAGAAGGGCccNNNNNNNNNNNNNNNNNNNNggGCCCTGAAGAAGGGCcc

PP7-RNA:

NNNNNNNNNNNNNNNNNNNNccggagcagacgatatggcgtcgctccgg

double hairpin RNA:

TGGAATAGTATAACAATATGCTAAATGTTGTTATAGTATCCCACNNNNNNNNNNNNNNNNNNNNGTGGAATAGTATAACAATATGCTAAATGTTGTTATAGTATCCCAC

A-U to G-C substitutions in AdRNA

v1:

v2:

v3:

v4:

v5:

v6:

v7:

v8:

v9:

v10:

v11:

v12:

v13:

dCas9 Cj-NES-linker-cdADAR2(E488Q)

Single ADAR2 recruitment domain and double ADAR2 recruitment domain: singly:

bis 1:

a double 2:

bis 3:

double 4:

double 5:

drawings

FIG. 1 is a schematic of a vector construct developed to deliver modified endogenous or orthogonal tRNA's.

FIGS. 2A-B show the suppression efficiency of tRNA constructs: (FIG. 2A) the relative efficiency of suppressor tRNA's derived from arginine, serine and leucine for amber, ochre and opal stop codons; representative images showing recovery of GFP expression in the presence of Ser tRNA amber; (FIG. 2B) comparison of the suppression efficiency of single or double pyrrolysinyl tRNA against amber, ochre and opal stop codons in the presence of 2mM UAA; representative images of relative GFP recovery using mono-and bispyrrolidinyl tRNA succinyls in the presence of 2mM UAA are shown.

Figure 3 shows GFP reporter results for dystrophin using various trnas and amino acids.

Figure 4 shows the results of dystrophin recovery experiments performed in mdx mice.

Figure 5 shows the sequence used to generate the ADAR2 construct.

Figure 6 shows a non-limiting example of RNA level point mutations that can be made to codons by ADAR 2.

Figure 7 shows an exemplary schematic of constructs that can be used in ADAR 2-based RNA editing systems.

FIG. 8 shows the optimization of the length of the adRNA and the distance of the editing zone from the ADAR2 recruitment domain. The first number in shorthand form for each category on the Y-axis is the length of the adRNA and the second number (behind the dashed line) is the distance of the editing zone from the ADAR2 recruitment domain. 20-6 with ADAR2 recruitment zone v2 gave us the best results.

Figure 9 shows the recovery of GFP expression in vitro using the editing system described herein.

Figure 10 shows the optimization results for hairpins with mismatches. The first number in the shorthand form of each class on the Y-axis is the number of mismatches and the second number is its number of bases from the target. For example, 13 is 1 mismatch, 3 bases from the target.

FIG. 11 shows the results for different lengths of the pivot guide RNA sequences without mismatches to the target.

Fig. 12A-C show the results of immunostaining (fig. 12A), western blot (fig. 12B), and in vitro OTC mRNA editing assay (fig. 12C).

Figure 13 is a western blot showing that the use of suppressor tRNA restored dystrophin expression compared to Cas 9-based methods.

Figure 14 shows normalized dystrophin mRNA levels.

Figure 15 shows the results of immunostaining.

FIGS. 16A-D show in vitro suppression and editing of stop codons in GFP reporter mRNA: (FIG. 16A) arginine, serine and leucine suppressor tRNA activity targeting amber, ochre and opal stop codons (n-3 independent replicates). (FIG. 16B) inhibition of amber, ochre and opal-type stop codons based on orthogonal tRNA/aaRS (MbpylRS) in the presence of one or two copies of pyrrollysyl tRNA delivered via AAV vector and in the presence of 1mM N ε -Boc-L-lysine (N ═ 3 independent replicates) (p values 0.022, 0.002, 0.027, respectively). (FIG. 16C) ADAR 2-based RNA editing efficiency for amber and ochre stop codons in one step, two steps, or in combination with suppressor tRNA (n-3 independent replicates). (fig. 16D) ADAR 2-based RNA editing efficiency (n ═ 3 or 6 independent replicates) for amber and ochre stop codons in the presence of one or two copies of adRNA delivered via AAV vectors (p values 0.0003, 0.0001, 0.0015, respectively).

Figures 17A-E show in vivo RNA targeting in a mouse model of human disease: (FIG. 17A) schematic representation of DNA and RNA targeting methods for restoring dystrophin expression in mdx mice: (i) a dual gRNA-CRISPR-based method that causes in-frame excision of exon 23; (ii) tRNA suppression of ochre-type codons; and (iii) ADAR 2-based edits to ochre-type codons. (FIG. 17B) immunofluorescence staining for dystrophin and nNOS in control and treated samples (scale bar: 250 μm). (figure 17C) in vivo TAA → TGG/TAG/TGA RNA editing efficiency in correspondingly treated adult mdx mice (n ═ 3 or 4 mice). (FIG. 17D) spf with G → A point mutation at the donor splice site or missense in the last nucleotide of exon 4ashSchematic representation of OTC locus in mice and methods to correct mutant OTC mRNA via ADAR2 mediated RNA editing. (FIG. 17E) adult spf at corresponding treatmentashIn vivo a → G RNA editing efficiency in mice (n ═ 3 or 4 mice).

FIGS. 18A-B show the evaluation and optimization of tRNA suppression in vitro: (FIG. 18A) specificity of modified serine suppressor tRNA for ochre-and opal-type stop codons (n-3 independent replicates). (FIG. 18B) three different aaRS were used: inhibition efficiencies of ochre-type stop codons for MbPylRS, mmphyrs, and AcKRS, as well as two or four copies of either pyrrolysinyltrna or serine suppressor tRNA, were all delivered using AAV vectors. MbpylRS: 1mM N epsilon-Boc-L-lysine; AcKRS: 1mM or 10mM N epsilon-acetyl-L-lysine (N ═ 3 independent replicates).

Figures 19A-C show evaluation and optimization of ADAR 2-mediated site-specific RNA editing in vitro: (FIG. 19A) GFP expression was restored when the adrA/radRNA had two mismatches corresponding to two adenosines in the ochre-type stop codon. The presence of a single mismatch leads to the formation of an amber or opal stop codon (n ═ 3 independent replicates). (FIG. 19B) one set of the adRNA designs used. (FIG. 19C) optimization of antisense region of the adRNA design 1 using the adRNA: length and distance from ADAR2 recruitment area were systematically varied, and editing efficiency was calculated as the ratio of sanger peak height G/(a + G) (n-3 independent replicates).

Figures 20A-C show targeting of dystrophin mRNA in vivo via suppressor tRNA: (FIG. 20A) the recovery of dystrophin expression gradually increased over time in mdx mice treated with AAV 8-bis-serine-ochre tRNA. (FIG. 20B) UAA-induced nNOS localization in mdx mice treated with AAV 8-bis-pyrrolysine-ochre type-tRNA-MbPylRS. (FIG. 20C) Western blot of dystrophin protein showed partial recovery of dystrophin expression in mdx mice treated with serine tRNA ochre, pyrrollysyl tRNA ochre and receiving UAA administration and in Cas9/gRNA treated samples.

Figures 21A-D show in vitro and in vivo editing of dystrophin and OTC mRNA: (FIG. 21A) representative Sanger sequencing charts showing 12.7% editing (using NGS quantification) of the ochre-type stop codon (TAA → TGG) in a fragment of mdx dystrophin mRNA expressed in HEK293T cells. (FIG. 21B) representative example of in vivo RNA editing analysis (using NGS quantification) of treated mdx mice. (FIG. 21C) representative Sanger sequencing charts showing spf expressed in HEK293T cellsash29.7% correction of point mutations in fragments of OTC mRNA (using NGS quantification). (FIG. 21D) treated spfashRepresentative examples of in vivo RNA editing analysis in mice (using NGS quantification).

FIGS. 22A-B show the in vitro editing efficiency of ADAR 2-E488Q. ADAR2-E488Q resulted in higher efficiency than ADAR2 in the in vitro editing of: (FIG. 22A) spf expressed in HEK293T cellsashFragments of OTC mRNA (n ═ 3 independent replicates) (p values: 0.037), and (fig. 22B) fragments of mdx dystrophin mRNA expressed in HEK293T cells (n ═ 3 independent replicates) (p values 0.048, 0.012, respectively). The efficiency was calculated as the ratio of the sanger peak height G/(a + G).

FIGS. 23A-D show (FIG. 23A) schematic representations of fusions of MCP or N22 with ADAR1 or ADAR 2; (FIG. 23B) schematic representation of ADRNA recruitment of APOBEC; (FIG. 23C) schematic representation of the more general construction of an adRNA; and (fig. 23D) schematic representation of the structure of the v2 adra scaffold after folding.

Figures 24A-B show schematic diagrams of optional embodiments in which (figure 24A) endogenous ADAR2 can be used in tissues with high endogenous ADAR2, e.g., brain, lung, and spleen, in the methods disclosed herein; and (fig. 24B) may increase ADAR1 and/or ADAR2 levels in tissues with low levels of endogenous ADAR1 and ADAR 2. Clockwise from the left, (1) delivery of adra and ADAR2 will result in high levels of RNA editing; (2) delivering adRNA alone may cause little or no editing due to low levels of endogenous ADAR1 and ADAR 2; (3) treatment of cells with IFN will cause an increase in the level of ADAR1(p150), but in the absence of adRNA, it is unlikely to cause any editing of the RNA target; (4) treatment of cells with IFN with the addition of adras will cause elevated levels of ADAR1(p150) and, in the presence of adras, may cause high levels of target RNA editing; (5) treatment of cells with IFN with the addition of adra and ADAR2 will result in elevated levels of ADAR1 expression and high levels of RNA editing.

Fig. 25 shows the conversion of UAA to UAG. UAA was converted to UAG via ADAR 2-based editing, and the addition of a suppressor tRNA targeting the UAG stop codon caused partial recovery of GFP expression.

Figure 26 shows the results of RNA editing in vivo in an mdx mouse model of muscular dystrophy.

Fig. 27 shows the resulting edit sequence resulting from the use of the promiscuous C-terminal ADAR 2.

Fig. 28 shows the editing efficiency of the stabilization bracket.

Figure 29 shows the scores of mRNA edited with single ADAR2 recruitment domain or double ADAR2 recruitment domain and the corresponding sequences.

FIG. 30 shows the scores of the mRNA edited in the case of using various MCP-ADAR scaffolds.

FIG. 31 shows alternative splice variants of OTC and are taken from Hodges, P.E. and Rosenberg, L.E.the spfresh mouse: a missense mutation in the ornithine transcarbamylase gene antigens mRNA spicing (missense mutations in the spfresh mouse: ornithine transcarbamylase gene also cause aberrant mRNA splicing), Proc.Natl.Acad.Sci.U.S.A.86,4142-4146 (1989).

Detailed Description

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All nucleotide sequences provided herein are presented in a 5 'to 3' orientation. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, devices, and materials are now described. All technical and patent publications cited herein are incorporated by reference in their entirety. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

The practice of the present technology will employ, unless otherwise indicated, conventional techniques of tissue culture, immunology, molecular biology, microbiology, cell biology, and recombinant DNA, which are within the skill of the art. See, e.g., Sambrook and Russell (2001), Molecular Cloning: A Laboratory Manual (Molecular Cloning: A Laboratory Manual), 3 rd edition; the book of Japan, Ausubel et al (2007), Current Protocols in molecular biology (recent Protocols in molecular biology); book Methods in Enzymology (Methods in Enzymology) (Academic Press, Inc., N.Y.); MacPherson et al (1991) PCR 1: A practical approach (PCR 1: practical methods) (IRL Press of Oxford University Press); MacPherson et al (1995) PCR 2: A Practical Approach (PCR 2: Practical methods); harlow and Lane, eds (1999) Antibodies, A Laboratory Manual (Antibodies: A Laboratory Manual); freshney (2005) Culture of Animal Cells: A Manual of Basic Technique (Animal cell Culture: Basic technical Manual), 5 th edition; gait, eds (1984) Oligonucleotide Synthesis (Oligonucleotide Synthesis); U.S. Pat. nos. 4,683,195; hames and Higgins eds (1984) Nucleic Acid Hybridization (Nucleic Acid Hybridization); anderson (1999) Nucleic Acid Hybridization (Nucleic Acid Hybridization); hames and Higgins, eds (1984) transformation and transformation; immobilised Cells and Enzymes (transcription and translation; Immobilized Cells and Enzymes) (IRL Press (1986)); perbal (1984) A practical guide to Molecular Cloning (Manual of practice for Molecular Cloning); miller and Calos (1987) Gene transfer Vectors for Mammalian Cells (Gene transfer Vectors for Mammalian Cells) (Cold Spring Harbor Laboratory); makrides, eds (2003) Gene Transfer and expression in Mammalian Cells (Gene Transfer and expression in Mammalian Cells); mayer and Walker, eds (1987) Immunochemical Methods in Cell and Molecular Biology (Immunochemical Methods in Cell and Molecular Biology) (Academic Press, London); and Herzenberg et al (1996) Weir's Handbook of Experimental Immunology (Handbook of Experimental Immunology by Well).

The terminology used in the description herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.

All numbers indicate, for example, that pH, temperature, time, concentration, and molecular weight (including ranges) are in approximate values varying (+) or (-) in increments of 1.0 or 0.1, suitably or alternatively +/-15%, or alternatively 10%, or alternatively 5%, or alternatively 2%. It should be understood that, although not always explicitly stated, the term "about" precedes all numerical indications. It is also to be understood that, although not always explicitly stated, the reagents described herein are exemplary only and that equivalents of these reagents are known in the art.

Unless the context indicates otherwise, it is specifically intended that the various features of the invention described herein can be used in any combination. Furthermore, the present disclosure also contemplates that, in some embodiments, any feature or combination of features described herein may be excluded or omitted. For purposes of illustration, if the specification states that a complex comprises components A, B and C, it is specifically intended that either one or a combination of A, B or C can be omitted and discarded, either alone or in any combination.

Unless expressly stated otherwise, all specified embodiments, features and terms are intended to include the recited embodiments, features or terms and their biological equivalents.

Definition of

As used in the specification and in the claims, the singular forms "a/an" and "the" include plural referents unless the context clearly dictates otherwise. For example, the term "a polypeptide" includes a plurality of polypeptides, including mixtures thereof.

The term "about" as used herein when referring to a measurable value such as an amount or concentration is meant to encompass a change of 20%, 10%, 5%, 1%, 0.5% or even 0.1% of the specified amount.

The term "comprising" as used herein is intended to mean that the compositions and methods include the recited elements, but do not exclude other elements. "consisting essentially of … …" when used in defining compositions and methods should be meant to exclude other elements having any substantial meaning for the combination intended for the intended use. Thus, a composition consisting essentially of elements as defined herein would not exclude trace contaminants from the isolation and purification processes and pharmaceutically acceptable carriers such as phosphate buffered saline, preservatives and the like. "consisting of … …" shall mean excluding more than trace amounts of other ingredient elements and substantial method steps for administering the compositions of the invention. Embodiments defined by each of these transitional terms fall within the scope of the present invention.

A "subject" for diagnosis or treatment is a cell or animal, such as a mammal or a human. Non-human animals that are diagnosed or treated are animals that are infected or animal models, such as simians; rats, such as rats, mice, chinchillas; canines, such as dogs; rabbits, such as rabbits; livestock, sport animals and pets.

The terms "protein", "peptide" and "polypeptide" are used interchangeably and in their broadest sense refer to a compound that is two or more subunit amino acids, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds. In another embodiment, the subunits may be linked by other linkages, such as esters, ethers, and the like. The protein or peptide must contain at least two amino acids and there is no limit to the maximum number of amino acids that can make up the sequence of the protein or the sequence of the peptide. The term "amino acid" as used herein refers to natural amino acids and/or unnatural amino acids or synthetic amino acids, including glycine as well as both D-and L-optical isomers, amino acid analogs, and peptidomimetics. The term "fusion protein" as used herein refers to a protein comprising domains from more than one naturally occurring or recombinantly produced protein, wherein in general each domain performs a different function. In this regard, the term "linker" refers to a protein fragment used to link these domains together, optionally maintaining the conformation of the fusion protein domains and/or preventing adverse interactions between the fusion protein domains that may impair their respective functions.

The terms "polynucleotide" and "oligonucleotide" are used interchangeably and refer to a polymeric form of nucleotides of any length (deoxyribonucleotides or ribonucleotides or analogs thereof). The polynucleotide may have any three-dimensional structure and may perform any known or unknown function. The following are non-limiting examples of polynucleotides: a gene or gene fragment (e.g., a probe, primer, EST, or SAGE tag), an exon, an intron, messenger RNA (mrna), transfer RNA, ribosomal RNA, RNAi, ribozyme, cDNA, recombinant polynucleotide, branched polynucleotide, plasmid, vector, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probe, and primer. Polynucleotides may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. Modifications to the nucleotide structure, if present, may be imparted before or after polynucleotide assembly. The sequence of nucleotides may be interrupted by non-nucleotide components. The polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The term also refers to double-stranded molecules and single-stranded molecules. Unless otherwise stated or required, any embodiment of the invention as a polynucleotide encompasses both the double-stranded form and each of the two complementary single-stranded forms known or predicted to make up the double-stranded form.

A polynucleotide consists of a specific sequence of four nucleotide bases: adenine (a); cytosine (C); guanine (G); thymine (T); and uracil (U) in place of thymine when the polynucleotide is RNA. In some embodiments, the polynucleotide may comprise one or more other nucleotide bases, such as inosine (I), a nucleoside formed when hypoxanthine is linked to ribofuranosyl via a β -N9-glycosidic bond, resulting in the following chemical structure:

inosine is read by the translation machinery as guanine (G). The term "polynucleotide sequence" is a letter representation of a polynucleotide molecule. The letter representation can be entered into a database in a computer with a central processing unit and used for bioinformatics applications such as functional genomics and homology searches.

As used herein, "expression" refers to the process by which a polynucleotide is transcribed into mRNA and/or the process by which transcribed mRNA is subsequently translated into a peptide, polypeptide, or protein. If the polynucleotide is derived from genomic DNA, expression may include splicing of mRNA in eukaryotic cells.

The terms "equivalent" or "bioequivalence" are used interchangeably when referring to a particular molecule, organism, or cellular material and are meant to refer to those having minimal homology while still maintaining the desired structure or function.

The term "encoding" as applied to a polynucleotide refers to a polynucleotide that is said to "encode" a polypeptide if it is in its native state, or which can be transcribed and/or translated to produce mRNA for the polypeptide and/or fragments thereof when manipulated by methods well known to those skilled in the art. The antisense strand is the complement of such a nucleic acid, and the coding sequence can be deduced therefrom.

The term "functional" as used herein may be used to modify any molecular, biological or cellular material to mean that it achieves a particular specified effect.

The terms "treating", "treatment", and the like, as used herein, are used herein to mean obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic in terms of completely or partially preventing a disease, disorder or condition or a sign or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse effects attributable to the disorder.

"administration" may be carried out in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective mode and dosage of administration are known to those skilled in the art and will vary with the composition used for treatment, the purpose of the treatment, the target cells being treated, and the subject being treated. Single or multiple administrations can be carried out using dose levels and patterns selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. The route of administration can also be determined and the method of determining the most effective route of administration is known to those skilled in the art and will vary with the composition used for treatment, the purpose of the treatment, the health or disease stage of the subject being treated and the target cell or tissue. Non-limiting examples of routes of administration include oral administration, nasal administration, injection, and topical administration.

The term "effective amount" refers to an amount sufficient to achieve the desired effect. In the context of therapeutic or prophylactic use, an effective amount will depend on the type and severity of the condition in question and the characteristics of the individual subject, such as general health, age, sex, body weight and tolerance to pharmaceutical compositions. In the context of immunogenic compositions, in some embodiments, an effective amount is an amount sufficient to elicit a protective response against a pathogen. In other embodiments, an effective amount of an immunogenic composition is an amount sufficient to cause the production of antibodies to an antigen. In some embodiments, an effective amount is the amount required to confer passive immunity to a subject in need thereof. With respect to immunogenic compositions, in some embodiments, an effective amount will depend on the intended use, the degree of immunogenicity of a particular antigenic compound, and the health/responsiveness of the subject's immune system, in addition to the factors described above. One skilled in the art will be able to determine the appropriate amount based on these and other factors.

In the context of in vitro applications, in some embodiments, an effective amount will depend on the size and nature of the application in question. It will also depend on the nature and sensitivity of the target in vitro and the method used. One skilled in the art will be able to determine an effective amount based on these and other considerations. According to embodiments, an effective amount may include one or more administrations of the composition.

The term "Cas 9" refers to the CRISPR-associated endonuclease referred to by this name (e.g., UniProtKB G3ECR1(Cas9_ STRTR)) as well as inactive Cas9 or dCas9 (e.g., having mutations in both the RuvC domain and the HNH domain) that lack endonuclease activity. The term "Cas 9" may further refer to an equivalent of the referenced Cas9 that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical thereto, including but not limited to other large Cas9 proteins. In some embodiments, Cas9 is derived from campylobacter jejuni (campylobacter jejuni) or another Cas9 ortholog having a length of 1000 amino acids or less.

The term "vector" refers to a polynucleotide (usually DNA) used to artificially carry foreign genetic material into another cell where it can be replicated or expressed. Non-limiting exemplary vectors include plasmids, viral vectors, cosmids, and artificial chromosomes. Such vectors may be derived from a variety of sources, including bacterial and viral sources. A non-limiting exemplary viral source of the plasmid is adeno-associated virus.

The term "recombinant expression system" as used herein refers to one or more genetic constructs for the expression of certain genetic material formed by recombination; in this regard, the term "construct" is interchangeable with the term "vector" as defined herein.

The term "adeno-associated virus" or "AAV" as used herein refers to a member of a class of viruses that are related to this name and belong to the Parvoviridae (parvoridae) dependent parvovirus genus (dependendopartial viruses). Various serotypes of this virus are known to be suitable for gene delivery; all known serotypes can infect cells from a variety of tissue types. At least 11 of the sequential numbering are disclosed in the prior art. Non-limiting exemplary serotypes that can be used for the purposes disclosed herein include any of the 11 serotypes, such as AAV2 and AAV 8.

The term "lentivirus" as used herein refers to a member of a class of viruses associated with this name and belonging to the genus lentivirus (lentivirus) of the family Retroviridae (Retroviridae). While some lentiviruses are known to cause disease, other lentiviruses are known to be suitable for gene delivery. See, for example, Tom-s et al (2013) Biochemistry, Genetics and Molecular Biology (Biochemistry, Genetics and Molecular Biology): "Gene Therapy-Tools and Potential Applications", ISBN 978-953-51-1014-9, DOI: 10.5772/52534.

The term "restore" with respect to protein expression as used herein refers to the ability to establish full-length protein expression, wherein the previous protein expression was truncated due to the mutation.

The term "mutation" as used herein refers to an alteration of a nucleic acid sequence encoding a protein relative to the consensus sequence of said protein. A "missense" mutation causes one codon to be substituted for another; a "nonsense" mutation changes a codon from that encoding a particular amino acid to a stop codon. Nonsense mutations often cause the translation of the protein to be truncated. A "silent" mutation is a mutation that has no effect on the resulting protein. The term "point mutation" as used herein refers to a mutation that affects only one nucleotide in a gene sequence. "splice site mutations" are those present in the precursor mRNA (prior to processing to remove introns) that cause mis-translation of the protein and often truncation of the protein due to incorrect delineation of the splice site.

"messenger RNA" or "mRNA" is a nucleic acid molecule that is transcribed from DNA and then processed to remove non-coding portions called introns. The resulting mRNA is exported from the nucleus (or another location where DNA is present) and translated into protein. The term "pre-mRNA" refers to the strand prior to processing to remove the non-coding portion.

A "transfer ribonucleic acid" or "tRNA" is a nucleic acid molecule that facilitates translation of an mRNA into a protein. tRNA has a unique folding structure, comprising three hairpin loops; one of these loops contains a "stem" portion that encodes an anti-codon. The anti-codon recognizes the corresponding codon on the mRNA. Each tRNA "carries" an amino acid corresponding to a codon of mRNA; this "carry over" is achieved by the enzyme tRNA synthetase. When the tRNA recognizes the codon corresponding to its anticodon, it transfers the amino acid it carries to the growing amino acid chain to form a polypeptide or protein. Endogenous tRNA's can be carried by endogenous tRNA synthetases. Thus, endogenous trnas typically carry canonical amino acids. An orthogonal tRNA derived from an external source requires a corresponding orthogonal tRNA synthetase. Such orthogonal tRNAs can have canonical amino acids and non-canonical amino acids. In some embodiments, the amino acid carried by the tRNA can be detectably labeled to enable detection in vivo. Techniques for labeling are known in the art and include, but are not limited to, click chemistry, wherein an azide/alkyne-containing unnatural amino acid is added by an orthogonal tRNA/synthetase pair, and thus, detection can be performed using an alkyne/azide-containing fluorophore or other such molecule.

The term "stop codon" means a trinucleotide sequence within the messenger RNA that signals the termination of translation. Non-limiting examples include UAG, UAA, UGA in RNA and TAG, TAA or TGA in DNA. Unless otherwise specified, the term also includes nonsense mutations within the DNA or RNA that introduce a premature stop codon, thereby causing any resulting protein to be abnormally shortened. Trnas corresponding to various stop codons are known under specific names: amber (UAG), ochre (UAA) and opal (UGA).

"canonical amino acids" refer to those 20 amino acids naturally occurring in humans, which are shown in the following table by their respective three-letter abbreviations, one-letter abbreviations, structures, and corresponding codons: non-polar aliphatic residue

Aromatic radicals

Polar uncharged residues

Positively charged residues

Negatively charged residues

The term "non-canonical amino acid" refers to those synthetic or otherwise modified amino acids that fall outside of this group, typically produced by chemical synthesis or modification of a canonical amino acid (e.g., amino acid analogs). The present disclosure uses protein non-canonical amino acids in some of the methods and vectors disclosed herein. A non-limiting exemplary non-canonical amino acid is pyrrolysine (Pyl or O), the chemical structure of which is provided below:

inosine (I) is another exemplary non-canonical amino acid that is commonly found in trnas and is necessary for proper translation according to "wobble base pairing. The structure of inosine is provided above.

The term "ADAR" as used herein refers to an adenosine deaminase that can convert adenosine (a) to inosine (I) in an RNA sequence. ADAR1 and ADAR2 are two exemplary ADARs involved in mRNA editing in vivo. Non-limiting exemplary sequences of ADAR1 may be found under the following reference numbers: HGNC: 225, a step of mixing; entrez Gene: 103; ensembl: ENSG 00000160710; OMIM: 146920, respectively; UniProtKB: p55265; and GeneCards: GC01M154554 and its bioequivalents. Non-limiting exemplary sequences of ADAR2 may be found under the following reference numbers: HGNC: 226; entrez Gene: 104; ensembl: ENSG 00000197381; OMIM: 601218, respectively; UniProtKB: p78563; and GeneCards: GC21P045073 and its bioequivalents. Additional non-limiting exemplary sequences of catalytic domains are provided above. The forward and reverse RNAs used to direct site-specific ADAR editing are referred to as "adRNA" and "radRNA," respectively. The catalytic domains of ADAR1 and ADAR2 are included in the sequences provided below.

ADAR1 catalytic domain:

KAERMGFTEVTPVTGASLRRTMLLLSRSPEAQPKTLPLTGSTFHDQIAMLSHRCFNTLTNSFQPSLLGRKILAAIIMKKDSEDMGVVVSLGTGNRCVKGDSLSLKGETVNDCHAEIISRRGFIRFLYSELMKYNSQTAKDSIFEPAKGGEKLQIKKTVSFHLYISTAPCGDGALFDKSCSDRAMESTESRHYPVFENPKQGKLRTKVENGEGTIPVESSDIVPTWDGIRLGERLRTMSCSDKILRWNVLGLQGALLTHFLQPIYLKSVTLGYLFSQGHLTRAICCRVTRDGSAFEDGLRHPFIVNHPKVGRVSIYDSKRQSGKTKETSVNWCLADGYDLEILDGTRGTVDGPRNELSRVSKKNIFLLFKKLCSFRYRRDLLRLSYGEAKKAARDYETAKNYFKKGLKDMGYGNWISKPQEEKNFYLCPV

ADAR2 catalytic domain:

QLHLPQVLADAVSRLVLGKFGDLTDNFSSPHARRKVLAGVVMTTGTDVKDAKVISVSTGTKCINGEYMSDRGLALNDCHAEIISRRSLLRFLYTQLELYLNNKDDQKRSIFQKSERGGFRLKENVQFHLYISTSPCGDARIFSPHEPILEEPADRHPNRKARGQLRTKIESGEGTIPVRSNASIQTWDGVLQGERLLTMSCSDKIARWNVVGIQGSLLSIFVEPIYFSSIILGSLYHGDHLSRAMYQRISNIEDLPPLYTLNKPLLSGISNAEARQPGKAPNFSVNWTVGDSAIEVINATTGKDELGRASRLCKHALYCRWMRVHGKVPSHLLRSKITKPNVYHESKLAAKEYQAAKARLFTAFIKAGLGAWVEKPTEQDQFSLT

the double stranded RNA binding domain (dsRBD) of ADAR is comprised in the sequences provided below.

ADAR dsRBD:

MDIEDEENMSSSSTDVKENRNLDNVSPKDGSTPGPGEGSQLSNGGGGGPGRKRPLEEGSNGHSKYRLKKRRKTPGPVLPKNALMQLNEIKPGLQYTLLSQTGPVHAPLFVMSVEVNGQVFEGSGPTKKKAKLHAAEKALRSFVQFPNASEAHLAMGRTLSVNTDFTSDQADFPDTLFNGFETPDKAEPPFYVGSNGDDSFSSSGDLSLSASPVPASLAQPPLPVLPPFPPPSGKNPVMILNELRPGLKYDFLSESGESHAKSFVMSVVVDGQFFEGSGRNKKLAKARAAQSALAAIFN

It will be appreciated that additional mutations may be made to the sequence of ADAR and/or its various domains. For example, the applicant of the present invention has generated both the E488Q and E1008Q mutants of ADAR1 and ADAR2 and "promiscuous" variants of ADAR2 (resulting from C-terminal deletions). This "promiscuous" variant is so called because it shows a promiscuity in the edited reads with several a's near the target sequence, showing a conversion of a to G (validated at 2 different loci). The sequence of this variant is provided below.

"promiscuous" ADAR2 variant:

MLRSFVQFPNASEAHLAMGRTLSVNTDFTSDQADFPDTLFNGFETPDKAEPPFYVGSNGDDSFSSSGDLSLSASPVPASLAQPPLPVLPPFPPPSGKNPVMILNELRPGLKYDFLSESGESHAKSFVMSVVVDGQFFEGSGRNKKLAKARAAQSALAAIFNLHLDQTPSRQPIPSEGLQLHLPQVLADAVSRLVLGKFGDLTDNFSSPHARRKVLAGVVMTTGTDVKDAKVISVSTGTKCINGEYMSDRGLALNDCHAEIISRRSLLRFLYTQLELYLNNKDDQKRSIFQKSERGGFRLKENVQFHLYISTSPCGDARIFSPHEPILEEPADRHPNRKARGQLRTKIESGEGTIPVRSNASIQTWDGVLQGERLLTMSCSDKIARWNVVGIQGSLLSIFVEPIYFSSIILGSLYHGDHLSRAMYQRISNIEDLPPLYTLNKPLLSGISNAEARQPGKAPNFSVNWTVGDSAIEVINATTGKDELGRASRLCKHALYCRWMRVHGKVPSHLLRSKITKPNVYHESKLAAKEYQAAKARLFTAFIKAGLGAWVEKPTEQDQFSLTP*

without being bound by theory, a C-terminal deletion in ADAR1 may produce the same or similar effect.

The term "lacking" as used herein refers to a particular agent being below normal levels (physiologically acceptable levels). In the context of proteins, lacking refers to a full-length protein that is below normal levels.

The term "dystrophin" as used herein refers to a protein corresponding to that name and encoded by gene Dmd; non-limiting examples of which are found under UniProt reference numbers P11532 (for humans) and P11531 (for mice).

The term "ornithine transcarbamylase" or "OTC" as used herein refers to the protein corresponding to the name and encoded by gene OTC; non-limiting examples of which are found under UniProt reference numbers P00480 (for humans) and P11725 (for mice). OTC deficiency is an X-linked genetic disease that results in high ammonia concentrations in the blood. In some cases, OTC deficiency is caused by a G → a splice site mutation in the donor splice site of exon 4 that results in missplicing of the precursor mRNA. Such mutations result in the formation of proteins that are elongated or carry point mutations. The OTC protein level was reduced to 1/15-1/20. FIG. 31 (taken from Hodges, P.E. and Rosenberg, L.E. the spfresh mouse: a missense mutation in the ornithine transcarbamylase gene genes absotant mRNAs cloning (spfresh mice: missense mutation in the ornithine transcarbamylase gene also causes aberrant mRNA splicing), Proc.Natl.Acad.Sci.U.S.A.86,4142-4146(1989)) shows the alternative forms produced. The sequences are provided below:

OTC precursor mRNA (wild type):

OTC precursor mRNA (mutant):

OTC mRNA (mis-spliced, mutant):

OTC mRNA (correct splice, mutant):

OTC mRNA (correct splicing, wild-type):

as shown above, when mutations are present, the correct splice variant can be generated; however, such generation can lead to missense mutations, which can also lead to OTC deficiency.

The terms "hairpin", "hairpin loop", "stem loop" and/or "loop", used alone or in combination with a "motif, are used in the context of an oligonucleotide to refer to a structure formed in a single-stranded oligonucleotide when complementary sequences within the single strand base pair to form a region that is conformationally similar to a hairpin or loop, when read in the opposite direction.

The term "domain" as used herein refers to a specific region of a protein or polypeptide and is associated with a specific function. For example, "a domain associated with an RNA hairpin motif" refers to a domain of a protein that binds one or more RNA hairpins. Such binding may optionally be specific for a particular hairpin. For example, the M2 phage coat protein (MCP) is capable of specifically binding to specific stem-loop structures, including but not limited to the MS2 stem-loop. See, e.g., Peabody, D.S. "The RNAbindingsite of bacteriophage MS2 coat protein (RNA binding site of bacteriophage MS2 capsid protein)", EMBOJ.12(2):595-600 (1993); coriigan and Chubb, "Biophysical Methods in Cell Biology", Methods in Cell Biology (2015). Similarly, λ N22, referred to herein as the "N22 peptide," is capable of specifically binding to specific stem-loop structures, including but not limited to the BoxB stem-loop. See, e.g., Cilley and Williamson, "Analysis of bacterial N protein and peptide binding to boxBRNAuse polyaramide gel electrophoresis (PACE) for binding of phage N proteins and peptides to boxB RNA", RNA 3(1):57-67 (1997). Sequences of MCP and MS2 stem loops and N22 peptide and BoxB loop are provided above in the context of fusion proteins with ADAR (MCP, N22 peptide) and use in adRNA (MS2 stem loop, BoxB loop), respectively.

The term "APOBEC" as used herein refers to any protein and its equivalents that fall within the evolutionarily conserved cytidine deaminase family that is involved in mRNA editing (catalyzing the conversion of C to U). In some aspects, the term APOBEC refers to any one of APOBEC1, APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3E, APOBEC3F, APOBEC3G, APOBEC3H, APOBEC4, or their respective equivalents. Provided herein are non-limiting exemplary sequences of fusion proteins comprising one or more APOBEC domains fused to ADAR domains or to alternative domains to make them suitable for use in RNA editing systems. To this end, APOBEC can be considered as an equivalent of ADAR, which catalyzes editing, although achieved by different transformations. Thus, without being bound by theory, applicants of the present invention believe that all embodiments contemplated herein for use with ADAR-based editing systems may be suitable for use in an APOBEC-based RNA editing system.

The term "interferon" as used herein refers to a group of signaling proteins known to be associated with an immune response. In the context of the present application, the interferon of interest is one that causes enhanced expression of ADAR. The correlation between interferon alpha and ADAR1 is well known, and therefore, the present disclosure contemplates the use of interferon alpha as a means to increase endogenous ADAR1 expression. Commercial sources of isolated or recombinant interferon alpha include, but are not limited to, Sigma-Aldrich (Sigma-Aldrich), R & D systems (R & D Sytems), Ebol (Abcam), and Serimer Feishi Scientific. Alternatively, interferon alpha may be produced using known vectors and given protein sequences, for example Q6QNB6 (human IFNA).

Where not explicitly recited and unless otherwise intended, when the disclosure relates to a polypeptide, protein, polynucleotide or antibody, an equivalent or bioequivalence thereof is intended to fall within the scope of the disclosure. The term "biological equivalents thereof" as used herein is intended to be synonymous with "equivalents thereof" when referring to a reference protein, antibody, polypeptide or nucleic acid, and means those having minimal homology while still maintaining the desired structure or function. Unless specifically recited herein, it is contemplated that any polynucleotide, polypeptide, or protein referred to herein also includes equivalents thereof. For example, an equivalent means having at least about 70% homology or identity, or at least 80% homology or identity, and optionally, or at least about 85%, or alternatively at least about 90%, or alternatively at least about 95%, or alternatively 98% percent homology or identity to a reference protein, polypeptide, or nucleic acid and exhibiting substantially equivalent biological activity to the reference protein, polypeptide, or nucleic acid. Alternatively, when referring to a polynucleotide, the equivalent is a polynucleotide that hybridizes under stringent conditions to the reference polynucleotide or its complement.

Applicants of the present invention have provided herein polypeptide and/or polynucleotide sequences for use in gene and protein transfer and expression techniques described below. It will be appreciated that the sequences provided herein may be used to provide substantially identical sequences that express the product and produce proteins with the same biological properties, although this is not always explicitly indicated. These "bioequivalent" or "biologically active" polypeptides are encoded by equivalent polynucleotides as described herein. When compared using a sequence identity method run under default conditions, they may have a primary amino acid sequence that is at least 60%, or alternatively at least 65%, or alternatively at least 70%, or alternatively at least 75%, or alternatively at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95%, or alternatively at least 98% identical to the reference polypeptide. Specific polypeptide sequences are provided as examples of specific embodiments. The sequence is modified by substituting amino acids with substitute amino acids having similar charges. Furthermore, an equivalent polynucleotide is a polynucleotide that hybridizes under stringent conditions to a reference polynucleotide or its complement, or, in the case of a polypeptide, a polypeptide encoded by a polynucleotide that hybridizes under stringent conditions to a reference encoding polynucleotide or its complementary strand. Alternatively, the equivalent polypeptide or protein is a polypeptide or protein expressed from an equivalent polynucleotide.

"hybridization" refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing (Watson-Crick base pairing), Hodgstein binding (Hoogstein binding), or in any other sequence-specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. Hybridization reactions can constitute a step in a wider process, such as the initiation of a PC reaction or the enzymatic cleavage of a polynucleotide by ribozymes.

Examples of stringent hybridization conditions include: an incubation temperature of about 25 ℃ to about 37 ℃; a hybridization buffer concentration of about 6 XSSC to about 10 XSSC; formamide concentrations of about 0% to about 25%; and a wash solution of about 4 XSSC to about 8 XSSC. Examples of moderate hybridization conditions include: an incubation temperature of about 40 ℃ to about 50 ℃; a buffer concentration of about 9 XSSC to about 2 XSSC; formamide concentrations of about 30% to about 50%; and a wash solution of about 5 XSSC to about 2 XSSC. Examples of high stringency conditions include: an incubation temperature of about 55 ℃ to about 68 ℃; a buffer concentration of about 1 XSSC to about 0.1 XSSC; formamide concentrations of about 55% to about 75%; and a wash solution of about 1 XSSC, 0.1 XSSC, or deionized water. Generally, the hybridization incubation time is 5 minutes to 24 hours, 1, 2, or more washing steps are used, and the washing incubation time is about 1 minute, 2 minutes, or 15 minutes. SSC is 0.15M NaCl and 15mM citrate buffer. It should be understood that equivalents of SSCs using other buffering systems may be employed.

"homology" or "identity" or "similarity" refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing the position in each sequence that can be aligned for comparison purposes. When a position in the compared sequences is occupied by the same base or amino acid, then the molecules are homologous at that position. The degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated" or "non-homologous" sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of the invention.

Embodiments of the present disclosure

Point mutations underlie many genetic diseases. In this regard, while programmable DNA nucleases have been used to repair mutations, their use for gene therapy presents a number of challenges: one is that the efficiency of homologous recombination is generally low in cells; secondly, the active nuclease has the risk of introducing permanent off-target mutation; and thirdly, the ubiquitous programmable nucleases often contain elements of non-human origin, thereby increasing the potential for immunogenicity in vivo. In view of these, methods that instead target RNA directly and use the molecular machinery native to the host would be highly desirable. To this end, the applicant of the present invention has engineered and optimized two complementary approaches, herein collectively referred to as tRiAD, based on the use of tRNA in codon suppression and adenosine deaminase in RNA editing. Specifically, by delivering modified endogenous tRNA or RNA editing enzyme ADAR and related guide RNA (adrna) via adeno-associated virus, applicants of the present invention enabled read-through and correction of the premature stop codon in an mdx mouse model of muscular dystrophy that carries a nonsense mutation in the dystrophin gene. Furthermore, applicants of the present invention engineered ADAR 2-mediated spf deficient in Ornithine Transcarbamylase (OTC)ashCorrection of point mutations in liver RNA in the mouse model. Taken together, the results disclosed herein establish the use of inhibitory tRNA and ADAR2 for RNA targeting in vivo, and this integrated tr ad approach is robust, genomically traceless, and potentially non-immunogenic because it utilizes effector RNA and human proteins.

Aspects of the present disclosure relate to a tRNA-based protein editing system, optionally alone or in combination with an ADAR-based RNA editing system, for a subject, comprising one or more forward guide RNAs ("adrnas") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR, wherein the ADAR-based RNA editing system specifically edits a point mutation in an RNA sequence encoding a gene.

A tRNA-based protein editing system can comprise an endogenously modified tRNA and/or an orthogonal tRNA to prevent off-target editing of a protein. In this regard, systems for controlling these trnas are disclosed below.

The construction of adrnas for use in ADAR-based RNA editing systems is relatively simple, comprising an RNA targeting domain complementary to the target and optionally one or two recruitment domains (also known as aptamers) that recruit the RNA binding domains of various proteins. The optional recruitment domain is located at the 5 'end and/or the 3' end of the RNA targeting domain. A schematic of the adRNA bound to its mRNA target is provided in FIG. 23C. In some embodiments, the adRNA has an a-C mismatch that facilitates the editing function of ADAR. Similar frameworks can be used to target pre-mRNA prior to intron processing by tailoring the scaffold to target pre-mRNA present in the nucleus. This approach is employed in a non-limiting exemplary approach involving OTC deficiency (involving splice site mutations), while mRNA editing approaches are employed in a non-limiting exemplary approach involving dystrophin deficiency (involving nonsense mutations).

Applicants of the present invention tested a series of scaffolds shown in figure 19C to recruit the RNA binding domain of ADAR. The sequence provided in the figure represents the recruitment domain and italicized N represents the nucleotide complementary to the target.CIs a mismatch that promotes editing function. Sequences of different lengths and mismatch positions can be tested to determine the best adRNA for the desired target. For example, residues in the recruitment domain of an adRNA generated by the applicant of the present invention are modified (5'-3') as follows:

v1:

v2:

v3:

v4:

v5:

v6:

v7:

v8:

v9:

v10:

v11:

v12:

v13:

the structure of V2 after folding is as provided in fig. 23D. And the corresponding radRNA was generated as follows:

a schematic of the resulting pairing of the adRNA and the radRNA to the target mRNA is shown in FIG. 16C.

The applicant of the present invention also applied an alternative scaffold framework, using two ADAR recruitment domains (black font) on either side of the targeting domain, while altering the position of the C mismatch in the targeting domain (italic N).

These non-limiting exemplary scaffolds provide templates for engineering adrnas and radrnas for specific targets and can be optimized based on comparative efficacy studies performed according to the exemplary methods disclosed herein.

In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops.

Without being bound by theory, the applicants of the present invention believe that the double stranded RNA binding motif from ADAR may bind to several double stranded RNA sequences and thus may have possible off-target effects. To avoid such effects, applicants of the present invention contemplate the use of an exogenous protein domain to recognize an RNA hairpin motif in an adra. Both ADAR1 and ADAR2 consist of an RNA binding domain and a catalytic domain, which catalyzes the conversion of adenosine to inosine. The catalytic domain may be uncoupled from the RNA-binding domain. Our goal is to achieve high editing efficiency on targeted adenosines while reducing off-target effects, and therefore alternative RNA binding domains are being investigated. The applicant of the present invention has fused the catalytic domain of ADAR1 or ADAR2 to a different RNA binding domain, such as MCP, N22 or inactive CjCas9 (or other RNA targeting CRISPR, such as from SaCas9, CRISPR-Cas13, etc.). After addition of appropriate guide rna (adrna), the fusion protein is recruited to the target, further catalyzing the change of adenosine to inosine. In this case, the inactive CjCas9 (and by extension other CRISPRs) essentially serves as an RNA binding domain, which in turn can be tethered to an effector.

The domain is fused to an ADAR catalytic domain to produce an ADAR that specifically targets a particular adRNA comprising an RNA hairpin motif. For example, the applicant of the present invention has used the MS2 phage coat protein (MCP) to fuse with the catalytic domains of ADAR1 or ADAR2 and their corresponding mutants E488Q and E1008Q, while using the MS2 stem loop on RNA to recruit the fusion protein (fig. 23A). Similar to this system, the applicant of the present invention has also used the N22 peptide to fuse with the catalytic domain of ADAR1 or ADAR2 (and their mutants) while using the boxB aptamer to recruit the fusion protein. Thus, one or two copies of ADAR can be recruited based on the addition of a single or double hairpin (MS2/BoxB loop) (fig. 23A). PP7 hairpins are also contemplated to be used in the same manner.

Non-limiting framework sequences for recruitment of MCP-based fusion proteins are provided below, where the C mismatch may vary within the targeting domain (lower case letters indicate those linkers that help stabilize the underlined hairpin):

single recruitment domain (underlined):

two recruitment domains (underlined):

similar non-limiting framework sequences are provided for N22-based fusion proteins:

single recruitment domain (underlined):

two recruitment domains (underlined):

another approach is to associate a recruitment domain in the adRNA with Cas9 and couple the inactive Cas9 with the ADAR catalytic domain, providing the same specific recruitment. Providing a Cas 9-based fusion protein with a non-limiting framework sequence for recruitment:

psp dCas13a recruitment (mismatch position can vary)

CAACATTATCGGGGAGTTTTGACCTCCAAGGTGTTGNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNN

Cj dCas9 recruitment (mismatch position can vary)

NNNNNNNNNNNNNNNNNNNNNNgttttagtccctgaaaagggactaaaataaagagtttgcgggactctgcggggttacaatcccctaaaaccgcttttttt

APOBECs also have RNA editing function (fig. 23B). Thus, they may be used instead of or in addition to an ADAR-based editing system. For example, applicants of the present invention have formed fusions of MCP/N22 peptide with APOBEC to engineer targeting C → T RNA editing. Furthermore, the applicant of the present invention has fused the double stranded RNA binding domain (dsRBD) of ADAR2 to APOBEC, so that APOBEC can be recruited by adRNA.

In any of the embodiments disclosed herein, the addition of a Nuclear Localization Signal (NLS) to the fusion protein can help target the nuclear RNA (i.e., the pre-mRNA), while the addition of a Nuclear Export Signal (NES) to the fusion protein can help target the cytoplasmic RNA. This approach is useful when editing splice site mutations, which can lead to incorrect processing of introns in the precursor mRNA and, therefore, the production of incorrect mRNA for translation. OTC deficiency is an example where targeting of precursor mRNA using an adra scaffold may be useful because most aberrant OTC expression results from splice site mutations, resulting in truncated OTC proteins. Further addition of an RNA localization tag to the adRNA will enable targeting of RNA in specific cellular compartments.

In further embodiments, wherein the adRNA comprises one or more RNA hairpin motifs, the one or more RNA hairpin motifs are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

More generally, it is understood that the RNA targeting domains of the adrnas are designed such that they are complementary to the target mRNA, but contain a C mismatch at the position of the target adenosine. The recruitment domain of the adRNA is constant. As non-limiting examples:

exemplary targets: OTC mRNA (mutation underlined)

ADRNA v2 (targeting domain length: 20bp, mismatch position after 6 bases):

ADRNA v2 (targeting domain length: 21bp, mismatch position after 6 bases):

radRNA v2 (targeting domain length: 20bp, mismatch position after 6 bases):

ADRNA bis (targeting domain length: 20bp, mismatch position after 5 bases, 14 bases):

ADRNA MS2 (targeting domain length: 20bp, mismatch position after 14 bases):

ADRNA MS2 bis (targeting domain length: 20bp, mismatch position after 5 bases, 14 bases):

AdRNA boxB (targeting domain length: 20bp, mismatch position after 14 bases):

AdRNA BoxB bis (targeting domain length: 20bp, mismatch position after 5 bases, 14 bases):

a concerted or alternative approach to prevent off-target effects is to use endogenous ADAR. ADAR2 is highly expressed in tissues such as brain, lung and spleen, while ADAR1 is ubiquitously expressed at levels higher than the general expression level of ADAR 1. Thus, applicants of the present invention propose two pathways to engineer RNA editing by endogenous ADAR. First, ADAR1 expression may be stimulated by molecules such as interferons, e.g., interferon alpha. Second, scaffolds can be specifically engineered for recruitment of ADAR1 and experiments were performed using the v1-v13 scaffold as well as some of the chemically modified scaffolds disclosed above. The use of endogenous ADAR, rather than overexpression, may help to limit off-target effects.

Recombinant expression systems and vectors

Aspects of the present disclosure relate to vectors and recombinant expression systems.

For example, some aspects relate to a vector encoding one or more trnas having an anticodon sequence that recognizes a codon that comprises a point mutation in an RNA sequence encoding a protein, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the vector is an AAV vector, optionally an AAV8 vector. In some embodiments, the protein is dystrophin.

A further aspect relates to a recombinant expression system comprising one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR for a subject, wherein the ADAR-based RNA editing system specifically edits a point mutation in an RNA sequence encoding a protein. In some embodiments, the point mutation produces a nonsense mutation, optionally a premature stop codon, having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

Generally, methods for packaging genetic material, such as RNA, into one or more vectors are well known in the art. For example, genetic material can be packaged using packaging vectors and cell lines and introduced via conventional recombinant methods.

In some embodiments, packaging vectors can include, but are not limited to, retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors (optionally AAV 8). The packaging vector contains elements and sequences that facilitate the delivery of genetic material into the cell. For example, the retroviral construct is a packaging plasmid comprising at least one retroviral helper DNA sequence derived from a non-replicating retroviral genome in trans encoding all the viral particle proteins required for packaging of a non-replicating retroviral vector and for the production of viral particle proteins at high titers capable of packaging of a non-replicating retroviral vector without the production of a replicating helper virus. The retroviral DNA sequence lacks regions encoding the native enhancer and/or promoter of the viral 5'LTR of the virus and lacks the psi functional sequences and 3' LTR responsible for packaging the helper genome, but encodes a foreign polyadenylation site, such as the SV40 polyadenylation site and a foreign enhancer and/or promoter that directs efficient transcription in the cell type in which production of the virus is desired. The retrovirus is a leukemia virus, such as Moloney Murine Leukemia Virus (MMLV), Human Immunodeficiency Virus (HIV), or Gibbon Ape Leukemia Virus (GALV). The extrinsic enhancer and promoter may be the Human Cytomegalovirus (HCMV) Immediate Early (IE) enhancer and promoter, the Moloney Murine Sarcoma Virus (MMSV) enhancer and promoter (region U3), the Rous Sarcoma Virus (RSV) region U3, the splenomegalogenic virus (SFFV) region U3, or the HCMVIE enhancer linked to the native Moloney Murine Leukemia Virus (MMLV) promoter.

A retroviral packaging plasmid may consist of two retroviral helper DNA sequences encoded by a plasmid-based expression vector, for example wherein the first helper sequence comprises cDNA encoding the gag and pol proteins of avidity MMLV or GALV and the second helper sequence comprises cDNA encoding the env protein. The Env genes that determine the host range may be derived from genes encoding heterophilic, amphotropic, avitropic, polyhalotropic (mink focalization) or 10a1 murine leukemia virus Env protein or Gibbon Ape Leukemia Virus (GALV) Env protein, human immunodeficiency virus Env (gp160) protein, Vesicular Stomatitis Virus (VSV) G protein, human T cell leukemia (HTLV) Env gene products of class I and II, chimeric envelope genes derived from combinations of one or more of the above Env genes or chimeric envelope genes encoding the cytoplasm and transmembrane regions of the above Env gene products and monoclonal antibodies directed against specific surface molecules on the desired target cells. Similar vector-based systems may employ other vectors, such as sleeping beauty (sleeping beauty) vectors or transposon elements.

The resulting packaged expression system can then be introduced via an appropriate route of administration, as discussed in detail in connection with the methods disclosed herein.

Composition comprising a metal oxide and a metal oxide

A further aspect relates to a composition comprising any one or more of the vectors disclosed herein. In some embodiments, the composition further comprises an effective amount of interferon to enhance endogenous ADAR1 expression. In yet further embodiments, the interferon is interferon alpha.

Briefly, a pharmaceutical composition of the present disclosure, including but not limited to any of the claimed compositions, can comprise a target cell population as described herein in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients.

Examples of well-known carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, agarose, and magnetite. For the purposes of this disclosure, the nature of the carrier may be soluble or insoluble. Those skilled in the art will know of other suitable carriers for binding the antibody, or will be able to determine such carriers using routine experimentation.

Such compositions may also comprise buffering agents, such as neutral buffered saline, phosphate buffered saline, and the like; carbohydrates, such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids, such as glycine; an antioxidant; chelating agents, such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The compositions of the present disclosure may be formulated for oral, intravenous, topical, enteral, and/or parenteral administration. In certain embodiments, the compositions of the present disclosure are formulated for intravenous administration.

Administration of the composition may be carried out in a single dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective mode of administration and dosage are known to those skilled in the art and will vary with the composition used for treatment, the purpose of the treatment, and the subject being treated. Single or multiple administrations can be carried out using dose levels and patterns selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. In another aspect, the cells and compositions of the present disclosure may be administered in combination with other therapies.

The vectors, recombinant expression systems, and/or compositions are administered to the host using methods known in the art. Such administration of the compositions of the present disclosure can be performed to generate animal models of desired diseases, disorders, or conditions for experimental and screening assays.

Briefly, a pharmaceutical composition of the present disclosure, including but not limited to any of the claimed compositions, may comprise one or more vectors or recombinant expression systems as described herein in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients. Such compositions may comprise buffering agents, such as neutral buffered saline, phosphate buffered saline, and the like; carbohydrates, such as glucose, mannose, sucrose or dextran, mannitol; a protein; polypeptides or amino acids, such as glycine; an antioxidant; chelating agents, such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and a preservative. The compositions of the present disclosure may be formulated for oral, intravenous, topical, enteral, and/or parenteral administration. In certain embodiments, the compositions of the present disclosure are formulated for intravenous administration.

The pharmaceutical compositions of the present disclosure may be administered in a manner suitable for the disease, disorder or condition to be treated or prevented. The amount and frequency of administration will be determined according to factors such as the condition of the patient and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.

Method for restoring protein expression

Aspects of the present disclosure relate to methods of restoring protein expression.

For example, some aspects of the disclosure relate to a method of restoring expression of a protein comprising a point mutation in an RNA sequence encoding the protein in a subject in need thereof, the method comprising administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In further embodiments, pyrrolysine is introduced into the subject's diet. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the protein is dystrophin.

Other aspects relate to a recombinant expression system comprising one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR for a subject, wherein the ADAR-based RNA editing system specifically edits a point mutation in an RNA sequence encoding a protein. In some embodiments, the point mutation produces a nonsense mutation, optionally a premature stop codon, having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, optionally in embodiments involving nonsense or missense mutations, the targeted RNA is mRNA. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the protein is dystrophin. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In some embodiments, optionally in embodiments involving splice site mutations, the targeted RNA is a precursor mRNA. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA.

In either case, the assessment of whether protein expression "recovers" is achieved by any means of protein quantification when compared to baseline. The baseline may optionally be adjusted for the age, ethnicity, and other relevant demographic information of the subject based on previous levels of the subject or calculated as normal levels in the population. Techniques for quantifying protein expression are well known in the art and may optionally utilize controls or thresholds for comparison to a baseline value. Methods known in the art for these studies include, but are not limited to, qRTPCR, ELISA, western blotting, western immunostaining, spectroscopic and/or spectrometry based methods, and other assays commonly performed to determine the amount of protein expression in a sample from a subject. Alternatively, the "recovery" effect may be determined based on clinical outcome. For example, abnormal dystrophin levels are associated with symptoms of muscular dystrophy. Thus, recovery of expression can be determined exogenously based on clinical signs, such as the alleviation or reversal of these symptoms. For dystrophin, improvement in muscle strength may be one such indicator. Thus, the physician can perform force measurements to determine the result. Another example is Ornithine Transcarbamylase (OTC); abnormal OTC levels are the result of rare X-linked genetic disorders that result in excessive accumulation of ammonia in the blood (due to nitrogen accumulation). Thus, a relevant clinical outcome would be a reduction in ammonia in a biological sample such as blood or urine. Similarly, the clinical signal associated with a protein downstream of a protein of interest and the expression of the protein may be relevant indicators of "recovery", wherein the protein of interest is involved in a particular pathway.

Method of treatment

Point mutations are implicated in a number of diseases, disorders and conditions. Non-limiting examples are provided in table 1 below.

TABLE 1

Additional non-limiting examples include ornithine transcarbamylase deficiency, Nougarte night blindness (Nougaret night blindness), Uuscle's syndrome, atrial fibrillation, Duchenne muscular dystrophy, Wilson's disease, hereditary tyrosinemia, and some cancers that carry an A → G mutation in a gene such as B-catenin.

Accordingly, aspects of the present disclosure relate to the treatment of certain diseases, disorders, and conditions involving point mutations.

For example, some method aspects relate to treating a disease, disorder, or condition in a subject in need thereof, characterized by the presence of a point mutation in an RNA sequence encoding a protein associated with the disease, disorder, or condition, the treatment comprising administering to the subject a vector encoding one or more trnas having an anticodon sequence that recognizes a codon comprising the point mutation, optionally wherein the point mutation produces a premature stop codon. In some embodiments, the point mutation produces a nonsense mutation having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the tRNA is an endogenous tRNA having a modified anticodon stem that recognizes a codon comprising the point mutation. In further embodiments, the tRNA carries serine. In some embodiments, the tRNA is an orthogonal tRNA with a non-canonical amino acid. In additional embodiments, the vector further comprises a corresponding tRNA synthetase. In some embodiments, the corresponding synthetase is an E.coli glutaminyl-tRNA synthetase. In some embodiments involving orthogonal trnas, the non-canonical amino acid is pyrrolysine. In further embodiments, pyrrolysine is introduced into the subject's diet. In some embodiments, the vector encodes two trnas having anticodon sequences that recognize codons that comprise the point mutations. In some embodiments, the disease, disorder or condition is selected from the group consisting of the diseases, disorders and conditions listed in table 1, optionally characterized by the presence of a nonsense mutation and/or a premature stop codon. In some embodiments, the protein is dystrophin. In additional embodiments, the disease, disorder, or condition is muscular dystrophy. In yet further embodiments, the disease, disorder, or condition is duchenne muscular dystrophy.

A further method aspect relates to a method of treating a disease, disorder or condition in a subject in need thereof, the disease, disorder or condition characterized by the presence of a point mutation in an RNA sequence encoding a protein associated with the disease, disorder or condition, the method comprising administering to the subject one or more vectors encoding an ADAR-based RNA editing system comprising one or more forward guide RNAs ("adrnas") for ADAR and one or more corresponding reverse guide RNAs ("radrnas") for ADAR, wherein the ADAR-based RNA editing system specifically edits the point mutation. In some embodiments, the point mutation produces a nonsense mutation, optionally a premature stop codon, having a DNA sequence TAA and an RNA sequence UAA. In some embodiments, the ADAR-based RNA editing system converts UAA to UIA and, optionally, further converts UIA to UII. In some embodiments, the ADAR-based RNA editing system converts UAA to UAI. In some embodiments, optionally in embodiments involving nonsense or missense mutations, the targeted RNA is mRNA. In additional embodiments, the one or more vectors further encode a tRNA that targets an amber codon. In some embodiments, the protein is dystrophin. In some embodiments, the point mutation produces a splice site or missense mutation having a DNA sequence CAG and an RNA sequence CAG. In some embodiments, the ADAR-based RNA editing system converts CAG to CIG. In some embodiments, optionally in embodiments involving splice site mutations, the targeted RNA is a precursor mRNA. In some embodiments, the ADAR-based editing system further comprises ADAR1, ADAR2, their respective E488Q and E100Q mutants, a fusion protein comprising the catalytic domain of ADAR and a domain associated with an RNA hairpin motif, a fusion protein comprising the catalytic domain of ADAR and inactive Cas9, or a fusion protein comprising the double-stranded binding domain of ADAR and APOBEC. In further embodiments, the domain associated with the RNA hairpin motif is selected from the group of MS2 phage capsid protein (MCP) and N22 peptide. In some embodiments, the method further comprises administering an effective amount of interferon to enhance endogenous ADAR1 expression. In yet further embodiments, the interferon is interferon alpha. In some embodiments, the adRNA comprises one or more RNA hairpin motifs. In some embodiments, the one or more RNA hairpin motifs are selected from the group of MS2 stem loops and BoxB loops and/or are stabilized by replacing a-U with G-C. In some embodiments, the adRNA is stabilized via the incorporation of one or more of 2' -O-methyl, 2' -O-methyl 3' phosphorothioate, or 2' -O-methyl 3' thiopace at either or both termini of the adRNA. In some embodiments, the disease, disorder, or condition is selected from the group consisting of the diseases, disorders, and conditions listed in table 1. In additional embodiments, the protein is dystrophin and the disease, disorder, or condition is muscular dystrophy. In yet further embodiments, the disease, disorder, or condition is duchenne muscular dystrophy.

One of ordinary skill in the art will appreciate that the dosage and route of administration employed in these methods may vary based on the subject and the disease, disorder or condition being treated. Based on the knowledge in the art, such suitable dosages and routes may be selected based on the age, ethnicity, and other relevant demographic factors of the subject.

Reagent kit

In a specific aspect, the present disclosure provides kits for performing any of the methods disclosed herein and instructions for performing the methods of the present disclosure and/or administering the vectors, recombinant expression systems, and compositions disclosed herein.

The kit may also contain reagents required to preserve those components contained therein, such as buffers, preservatives or protein stabilizers. The kit may also contain components required for the detection of a detectable label, such as an enzyme or substrate. The kit may also contain a control sample or a series of control samples, which may be assayed and compared to the test sample. Each component of the kit may be enclosed in a separate container and all of the various containers may be placed in a single package along with instructions for interpreting the results of the assays performed using the kit. The kits of the present disclosure may contain the written product on or in a kit container. The written product describes how to use the reagents contained in the kit.

The suggested kit components may be packaged in the manner customary to those skilled in the art, if applicable. For example, these suggested kit components may be provided in the form of a solution or liquid dispersion, or the like.

102页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于分离靶核酸的组合物和方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!