SELF-REGULATING AAV VECTORS FOR SAFE EXPRESSION OF MECP2 IN RETT SYNDROME

In some aspects, the disclosure relates to compositions and methods of engineering a transgene. In some embodiments, the disclosure provides self-regulating recombinant nucleic acids, viral vectors and pharmaceutical compositions comprising a MeCP2 transgene. In some embodiments, compositions and methods described by the disclosure are useful for treating diseases and disorders associated with a loss of function mutation, for example Rett syndrome.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. § 119(c) of the filing date of U.S. Provisional Application Ser. No. 62/516,060, filed on Jun. 6, 2017, the entire contents of which are incorporated herein by reference.

BACKGROUND

Rett syndrome is a neurological disease caused by loss of function mutations in MeCP2. It has been observed that post-natal restoration of MeCP2 expression is effective in reversing some of the phenotypes present in MeCP2 mice, but safety concerns remain. Additionally, studies examining the therapeutic efficacy of certain MeCP2 vectors encoding the e1isoform have observed only partial rescue of Rett phenotypes. These studies have typically focused on neonatal intravascular (IV) or intracerebroventricular (ICV) delivery, and in some instances have encountered lethal liver toxicity and hindlimb clasping.

SUMMARY

Aspects of the disclosure relate to the discovery that certain combinations of miRNA regulatory elements (MREs), for example miRNA binding sites associated with gene expression negative feedback loops and miRNA binding sites that de-target transgene expression from non-target tissues, enable tunable transgene expression within a narrow range compatible with normal protein function and avoidance of off-target transgene toxicity. In some embodiments, compositions and methods described by the disclosure are therefore useful for treating diseases and disorders associated with loss of function mutations, for example Rett syndrome which is associated with loss of function mutations in the MECP2 gene.

Accordingly, in some aspects, the disclosure provides a method of engineering a transgene, the method comprising: selecting a first gene encoding a first product in a cell selecting a second gene encoding a second product in the cell; determining that expression of the second product is positively regulated by the first product in the cell; selecting an miRNA; determining that expression of the miRNA is positively regulated by the second product in the cell; and, engineering a transgene to express in the cell a transcript having a coding region encoding the first product and having one or more binding sites for the miRNA.

In some aspects, the disclosure provides a method of engineering a transgene, the method comprising: selecting a first gene encoding a first product in a cell; selecting an miRNA, the expression of which is positively regulated by the first product in the cell; and, engineering a transgene that expresses a transcript having a coding region encoding the first product and a 3′-non-coding region comprising one or more binding sites for the miRNA.

In some aspects, the disclosure provides a method of engineering a transgene, the method comprising: selecting a first gene encoding a first product in a cell; selecting a second gene encoding a second product in the cell; determining that expression of the second product is positively regulated by the first product in the cell; selecting an miRNA; determining that expression of the miRNA is positively regulated by the second product in the cell; and, engineering a transgene to express in the cell a transcript having a coding region encoding the first product and a 3′-non-coding region comprising one or more binding sites for the miRNA.

In some aspects, the disclosure provides a recombinant nucleic acid encoding a transcript having i) a coding region encoding a protein and ii) two or more miRNA binding sites, wherein the two or more miRNA binding sites comprise: at least one first, miRNA binding site specific for a first miRNA that is positively regulated by expression of the protein in a cell of a target tissue; and at least one second miRNA binding site specific for a second miRNA that is expressed, independent of expression of the protein, in cells of a non-target tissue.

In some aspects, the disclosure provides a recombinant nucleic acid encoding a transcript having a coding region encoding human MeCP2 protein or a functional fragment thereof and a 3′-non-coding region comprising one or more miRNA binding sites, wherein the one or more miRNA binding sites comprise: at least one miRNA binding site specific for an miRNA that negatively regulates expression of the transcript; and at least one miRNA binding site specific for an miRNA that inhibits expression of the transcript in a non-target tissue.

In some aspects, the disclosure provides a recombinant nucleic acid encoding a transcript having: a coding region encoding human MeCP2 or a functional fragment thereof and, a 3′-non-coding region comprising one or more miRNA binding sites, wherein transcript is flanked by adeno-associated virus (AAV) inverted terminal repeats (ITRs). In some embodiments, an AAV ITR is an AAV2, AAV3, AAV4, AAV5, or AAV6 ITR. In some embodiments, AAV ITRs are AAV2 ITRs. In some embodiments, ITRs are artificial sequences that replace ITR function, for example as disclosed in WO/2016/17208).

In some aspects, the disclosure provides a viral vector comprising a recombinant nucleic acid as described by the disclosure. In some embodiments, a viral vector is an adeno-associated virus (AAV) vector, an adenovirus vector, a lentiviral vector, a herpesvirus vector, or a baculovirus vector.

In some aspects, the disclosure provides a recombinant adeno-associated virus (rAAV) comprising: a recombinant nucleic acid as described by the disclosure; at least one adeno-associated virus (AAV) inverted terminal repeat (ITR); and a capsid protein.

In some aspects, the disclosure provides a recombinant AAV (rAAV) vector for self-regulated expression of a protein, the rAAV vector comprising a nucleic acid engineered to express in a cell of a target tissue a transcript encoding the protein, wherein the transcript comprises at least one first miRNA binding site specific for a first miRNA, wherein expression of the first miRNA is positively regulated by expression of the protein in the cell.

In some aspects, the disclosure provides a composition comprising a recombinant nucleic acid as described by the disclosure, or an rAAV as described by the disclosure, and a pharmaceutically acceptable excipient. In some embodiments, a composition is formulated for injection, for example systemic injection (e.g., intravenous injection) or intrathecal injection.

In some embodiments, a first product is a protein. In some embodiments, the protein is MeCP2, for example MeCP2 isoform e1 or MeCP2 isoform e2. In some embodiments, a first product is an miRNA or a long non-coding RNA.

In some embodiments, a second product is a protein, or nucleic acid. In some embodiments, the second product is bone-derived neurotrophic factor (BDNF). In some embodiments, the nucleic acid is an miRNA (e.g., miR-132). In some embodiments, the first miRNA is miR-132.

In some embodiments, at least one miRNA binding site specific for an miRNA that negatively regulates expression of the transcript comprises a miR-132 binding site, for example two or three miR-132 binding sites.

In some embodiments, at least one miRNA binding site specific for an miRNA that inhibits expression of the transcript in a non-target tissue comprises a miR-1 binding site, mir-122 binding site, or miR-1 and miR-122 binding site. In some embodiments, the at least one miRNA binding site specific for an miRNA that inhibits expression of the transcript in a non-target tissue comprises three miR-1 binding sites (e.g., 3×-miR-1) and three niR-122 binding sites (e.g., 3×-miR-122).

In some embodiments, methods described by the disclosure further comprise the step of engineering the 3′-non-coding region of the transcript to comprise one or more binding sites for one or more de-targeting miRNAs. In some embodiments, one or more de-targeting miRNAs inhibit expression of the transgene from liver, heart, lung, muscle, pancreas, or immune (e.g., antigen presenting) cells. In some embodiments, one or more de-targeting miRNA is miR-122, miR-1, or miR-122 and miR-1. In some embodiments, one or more de-targeting miRNAs inhibit expression of the transgene in immune cells, such as antigen presenting cells (e.g., dendritic cells, macrophages, etc.). In some embodiments, one or more de-targeting miRNA is miR-15a, miR-16-1, miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, miR-21, miR-29a, miR-29b, miR-29c, miR-30b, miR-31, miR-34a, miR-92a-1, miR-106a, miR-125a, miR-125b, miR-126, miR-142-3p, miR-146a, miR-150, miR-155, miR-181a, miR-223 or miR-424.

In some embodiments, an miRNA binding site or miRNA binding sites is located between the last codon of the coding region and the poly-A tail of the transcript.

In some embodiments of methods described by the disclosure, the step of engineering the transgene comprises inserting the transgene into a vector. In some embodiments, a vector is a cloning vector, expression vector, plasmid, or viral vector.

In some embodiments, a recombinant nucleic acid further comprises a promoter, for example a mouse MeCP2 promoter. In some embodiments, a mouse MeCP2 promoter comprises the sequence set forth in SEQ ID NO: 3.

In some embodiments, a recombinant nucleic acid is located on a plasmid.

In some embodiments, a capsid protein is a capsid protein that facilitates crossing of the rAAV across the blood-brain barrier of a subject. In some embodiments, a capsid protein has a serotype selected from the group consisting of AAV-PHP.B. AAV1, AAV2. AAV2i8, AAV2.5, AAV5, AAV6, AAV8. AAVrh8. AAV9. AAVrh10, AAV-B1, AAV9.45A-String (e.g., AAV9.45-AS), AAV9.45Angiopep, AAV9.47-Angiopep. and AAV9.47-AS. AAV5, AAVrh39. AAVrh43, CAM130, and AAV9HR. In some embodiments, a capsid protein has a serotype as described in WO2015/127128. WO2016/054554, WO2016054557, or WO2016/065001. In some embodiments, a capsid protein comprises or consists of a sequence set forth in SEQ ID NO: 14 or 15 (e.g., AAV-PHP.B or AAV9).

In some aspects, the disclosure provides a method of treating Rett syndrome in a subject, the method comprising, administering to a subject having or suspected of having Rett syndrome an effective amount of: a recombinant nucleic acid as described by the disclosure; a rAAV as described by the disclosure; or, a composition as described by the disclosure.

In some embodiments, the subject is a human subject. In some embodiments, a subject is less than one year old. In some embodiments, a subject is characterized by a mutation in at least one copy of the MeCP2 gene, for example a loss of function mutation.

In some embodiments, a recombinant nucleic acid, rAAV or composition as described by the disclosure is administered by injection, for example systemic injection (e.g., intravenous injection) or intrathecal injection. In some embodiments, the administration results in the effective amount of the recombinant nucleic acid, rAAV or composition crossing the blood-brain barrier of a subject. In some embodiments, the administration results in a non-toxic level of MeCP2 expression in the brain of the subject.

BRIEF DESCRIPTION OF DRAWINGS

FIGS. 1A-1B show characterization of new AAV-MeCP2 vectors for safe and effective gene therapy in Rett syndrome. FIG. 1A shows a schematic depiction of a homeostatic mechanism of MeCP2 auto-regulation. FIG. 1B shows a schematic depiction of the structure of self-regulated AAV-MeCP2 vectors encoding human MeCP2-e1 with a myc tag under a mouse MeCP2 promoter (−223 to +56) and different microRNA recognition elements (e.g., miR-122/IT; miR-132T).

FIGS. 2A-2C show effective expression of AAV2-MeCP2 in HEK293T cells FIG. 2A shows MeCP2 expression measured by Western blot. FIG. 2B shows MeCP2 expression measured by a normalized protein expression assay (FIG. 211). FIG. 2C shows a toxicity profile of 293T cells transduced with AAV2-MeCP2 for four days at a dose of 30.00) gc/cell.

FIGS. 3A-3C show AAV2-MeCP2 expression in mouse cortical neurons. FIG. 3A shows mouse primary cortical neurons were transduced at AAV vector doses ranging from 1E3-1E5 vg/cell including AAV-GFP as a control. FIG. 3B shows Western blot analysis of hMeCP2-myc expression in neurons 5 days after infection with 3E4 vector genomes(dose)/cell. FIG. 3C shows miR-132 expression in response to AAV2-MeCP2 re-delivery.

FIGS. 4A-4C show representative data obtained from in vivo mouse experiments. FIG. 4A shows wild-type post-natal day 1 mice injected via the facial vein with AAV encoding the e1 isoform of human MeCP2 containing 0, 1×, 2×, or 3× miR-132 target sequences. Wild-type animals were euthanized 3 months following injection and whole brain, heart and liver tissue was subjected to total RNA extraction, cDNA synthesis and qRT-PCR using primers specific to the e1 isoform of human MeCP2. Data were normalized to AAV-MeCP2 containing 3×miR-132 target sequences, which was set to 1. FIG. 4B shows gene expression analysis of human MeCP2 isoform e1 in brain of wild-type mice following intracranial injection of AAV-MeCP2.

FIG. 4C shows gene expression analysis of human MeCP2 isoform e1 in brain of wild-type mice following intracranial injection of AAV-MeCP2.

FIG. 5 shows MeCP2 expression driven by constructs described by the disclosure is effectively de-targeted from heart and liver.

DETAILED DESCRIPTION

Aspects of the disclosure relate, in part, to AAV vectors capable of self-regulating transgene (e.g., MeCP2) expression levels to prevent overexpression related toxicity. In some embodiments, the self-regulating mechanism is based on the presence of multiple copies of a miRNA regulatory element (e.g., one or more miR-132 binding sites) in the 3′UTR of the transgene cassette. As described further in the Examples section. AAV vectors capable of self-regulating transgene expression, in some embodiments, have an improved efficacy and safety profile compared to other AAV vectors, for example AAV vectors comprising native transgene promoters only. It should be recognized that the observations described in the Examples section in the context of miR-132/MeCP2 constructs is applicable to other transgene expression constructs comprising binding sites of other miRs that regulate protein expression (e.g., through a negative feedback loop).

In some embodiments, delivery routes that are most likely to mediate global gene delivery to the CNS (e.g., systemic injection and intrathecal injection) are likely to result in high level transduction of peripheral organs where transgene (e.g., MeCP2) expression may become toxic. The disclosure is based, in part, on the recognition that combining miRNA regulatory elements (MREs), such as miRNA binding sites (e.g., miR-122 binding sites and miR-1 binding sites), with MREs associated with negative feedback loops regulating protein expression (e.g., miR-132 binding sites for MeCP2), simultaneously regulate transgene expression levels and de-target transgene expression in peripheral organs.

Accordingly in some aspects, the disclosure provides a method of engineering a transgene, the method comprising: selecting a first gene encoding a first product in a cell; selecting an miRNA, the expression of which is positively regulated by the first product in the cell; and, engineering a transgene that expresses a transcript having a coding region encoding the first product and one or more binding sites for the miRNA. In some embodiments, the one or more binding sites for the miRNA are located in a 3′-non-coding region of the transcript.

In some aspects, the disclosure provides a method of engineering a transgene, the method comprising: selecting a first gene encoding a first product in u cell; selecting a second gene encoding a second product in the cell; determining that expression of the second product is positively regulated by the first product in the cell; selecting an miRNA; determining that expression of the miRNA is positively regulated by the second product in the cell; and, engineering a transgene to express in the cell a transcript having a coding region encoding the first product and one or more binding sites for the miRNA. In some embodiments, the one or more binding sites for the miRNA are located in a 3′-non-coding region of the transcript.

As used herein, “engineering a transgene” refers to production (e.g., synthesis) of a recombinant nucleic acid using gene cloning techniques, such as polymerase chain reaction (PCR) restriction enzyme digestion, and in vitro nucleic acid ligation, for example as described in Sambrook et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press. Cold Spring Harbor. N.Y.

As used herein, a “product” or “gene product” refers to a nucleic acid (e.g., RNA transcript, dsRNA, miRNA, etc.), a peptide, protein, or polypeptide that is transcribed and/or translated from a nucleic acid (e.g., DNA or RNA) sequence. In some embodiments, a product is an RNA transcript comprising a protein coding region. In some embodiments, a protein coding region encodes a protein associated with a disease caused by a loss of function mutation (e.g., MeCP2). Additional examples of proteins associated with a disease caused by a loss of function mutation include but are not limited to tyrosinase (Tyrosinemia), lysosomal acid beta-galactosidase (GM1-gangliosidosis) beta-hexosaminidase A and B (Tay-Sach and Sandhoff disease), aspartoacylase (ASPA; Canavan disease). Aspartylglucosamininidase (Aspartylglucosaminuria), Palmitoyl protein thioesterase (Infantile Batten disease), tripeptidyl peptidase (Late infantile Batten disease), α-Galactosidase (Fabry disease), α-Fucosidase (Fucosidosis). Protective protein/cathepsin A (Galactosialidosis), β-Glucosidase (Gaucher disease). Galactosylceramidase (Globoid-cell leukodystrophy), α-Mannosidase (α-Mannosidosis), Arylsulfatase A (Metachromatic leukodystrophy), α-L-iduronidase (Mucopolysaccharidosis I). α-N-acetylglucosaminidase (Mucopolysaccharidosis IIIB). Arylsulfatase B (Mucopolysaccharidosis VI). β-Glucuronidase (Mucopolysaccharidosis VID. Acid sphingomyelinase (Nieman-Pick disease), α-Glucosidase (Pompe disease) and Acid lipase (Wolman disease), FOXG1 (FOXG1 Syndrome), CDKL5, N-GlyI, Glut-1 (De Vivo disease), etc.

In some embodiments, a product is an interfering nucleic acid, for example a miRNA that regulates expression or activity of a gene product.

In some embodiments, one product regulates gene expression or protein expression of a second product. Regulation of gene product expression or translation can be positive or negative. “Positive regulation” refers to an increase of gene expression or activity (e.g., as a result of the expression or activity of another gene product). “Negative regulation” refers to a decrease or inhibition of gene expression or activity (e.g., as a result of the expression or activity of another gene product through a feedback loop).

In some embodiments, gene products such as growth factors, transcription factors (e.g., as described in Wang et al. Nucleic Acids Res. 2010 January; 38 (Database issue): D119-D122), etc. are capable of regulating transgene expression or activity in a cell or subject. Examples of growth factors include neurotrophins, such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin 3, neurotrophin 4, glial derived neurotrophic factor (GDNF), ciliary neurotrophic factor (CNTF), fibroblast growth factors (FGF1 to 23), neurturin, insulin-like growth factor 1 (IGF-1), insulin-like growth factor 2 (IGF-2), etc.

In some embodiments, transgenes as described by the disclosure are engineered to comprise at least one miRNA regulatory element (e.g., miRNA binding site) that is associated with a gene expression regulatory loop (e.g., negative feedback loops, positive feedback loops, etc.). Generally, gene expression regulatory loops may be endogenous to a cell, or artificial (e.g., one or more elements of the feedback loop are provided along with a transgene). In one example of a negative feedback loop, expression of MeCP2 in a cell causes an increase of brain-derived neurotrophic factor (BDNF) in the cell, which in turn increases expression of miR-132, which in turn regulates MeCP2 expression (FIG. 1). It should be appreciated that, in some embodiments, the disclosure relates to positive feedback loops, which may be used to amplify transgene expression.

In some embodiments, transgenes as described by the disclosure are engineered to comprise at least one miRNA regulatory element (e.g., miRNA binding site) that de-targets expression of the transgene from one or more non-target tissues. As used herein, “non-target tissue” refers to a tissue (e.g., cells of a tissue) in which expression of the transgene is undesirable. For example, in some embodiments, overexpression of MeCP2 in a cell results in hepatic cytotoxicity; in that context, liver tissue (e.g., liver cells) are a non-target tissue. In some embodiments, a non-target tissue is liver (e.g., liver cells), heart (e.g., heart cells), pancreas (e.g., pancreatic cells), muscle (e.g., muscle cell), immune cell (e.g., antigen presenting cells, etc.), or any combination thereof.

As used herein, “target tissue” refers to a tissue (e.g., cells of a tissue) in which expression of a transgene is preferred relative to other tissues, such as non-target tissues. In some embodiments, a target tissue is CNS tissue (e.g., CNS cells, such as neurons). Non-limiting examples of CNS tissue include brain tissue (e.g., neurons, glial cells, etc.) and spinal cord tissue.

Generally, the one or more miRNA binding sites of a transcript encoded by a transgene are located in the 3′ untranslated region (3′UTR) of the transcript. In some embodiments, the one or more miRNA binding sites are located between the last codon of the coding region of the transcript and the poly-A tail of the transcript. However, it should be appreciated that, in some embodiments, one or more miRNA binding sites are located in a region other than the 3′UTR of the transcript, for example in an intron at the 5′-end of the transcript. The number of miRNA binding sites engineered into a transgene as described by the disclosure will vary depending upon the gene product encoded by the transgene, and may be determined empirically by a skilled artisan without an undue amount of experimentation. For example, in some embodiments a transgene as described by the disclosure comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA binding sites. In some embodiments a transgene as described by the disclosure comprises more than 10 (e.g., any integer between 11 and 100) miRNA binding sites. In some embodiments, a transgene as described by the disclosure comprises 3, 4, or 5 miRNA binding sites. The one or more miRNA binding sites may each bind the same miRNA, or different miRNA. In some embodiments, a transgene as described by the disclosure comprises one or more (e.g. 3) miR-122 binding site(s), one or more (e.g., 3) miR-1 binding site(s) and three miR-132 binding sites.

Recombinant Nucleic Acids

In some embodiments, a transgene as described by the disclosure is encoded by a recombinant nucleic acid. A “nucleic acid” sequence refers to a DNA or RNA sequence. In some embodiments, proteins and nucleic acids of the disclosure are isolated. As used hemin, the term “isolated” means artificially produced. As used herein with respect to nucleic acids, the term “isolated” means: (i) amplified in vitro by, for example, polymerase chain reaction (PCR); (ii) recombinantly produced by cloning; (iii) purified, as by cleavage and gel separation; or (iv) synthesized by, for example, chemical synthesis. An isolated nucleic acid is one which is readily manipulable by recombinant DNA techniques well known in the art. Thus, a nucleotide sequence contained in a vector in which 5′ and 3′ restriction sites are known or for which polymerase chain reaction (PCR) primer sequences have been disclosed is considered isolated but a nucleic acid sequence existing in its native state in its natural host is not. An isolated nucleic acid may be substantially purified, but need not be. For example, a nucleic acid that is isolated within a cloning or expression vector is not pure in that it may comprise only a tiny percentage of the material in the cell in which it resides. Such a nucleic acid is isolated, however, as the term is used herein because it is readily manipulable by standard techniques known to those of ordinary skill in the art. As used herein with respect to proteins or peptides, the term “isolated” refers to a protein or peptide that has been isolated from its natural environment or artificially produced (e.g., by chemical synthesis, by recombinant DNA technology, etc.).

The skilled artisan will also realize that conservative amino acid substitutions may be made to provide functionally equivalent variants, or homologs of the capsid proteins. In some aspects the disclosure embraces sequence alterations that result in conservative amino acid substitutions. As used herein, a conservative amino acid substitution refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made. Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references that compile such methods, e.g., Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989, or Current Protocols in Molecular Biology, F. M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York. Conservative substitutions of amino acids include substitutions made among amino acids within the following groups: (a) M, I, L, V; (b) F, Y, W; (c) K, R, H; (d) A, G; (e) S, T; (f) Q, N; and (g) E, D. Therefore, one can make conservative amino acid substitutions to the amino acid sequence of the proteins and polypeptides disclosed herein.

In some embodiments, a nucleic acid as described by the disclosure is contained within a vector. As used herein, the term “vector” includes any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, artificial chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors. Examples of viral vector include adenovirus vector, adeno-associated virus (AAV) vector, lentiviral vectors, herpesvirus vectors, baculovirus vectors, etc.

MeCP2

In some aspects, the disclosure relates to compositions and methods for expressing MeCP2 protein in a cell or subject. “MeCP2” refers to methyl CpG binding protein 2, which is encoded by the MeCP2 gene and plays important roles (e.g., functions as a transcriptional repressor, or transcriptional activator) in nerve cells, such as mature neurons. One example of a MeCP2 gene is represented by GenBank Accession Number NM_001110792 (MeCP2-e1). Another example of a MeCP2 gene is represented by GenBank Accession Number NM_001110792 (MeCP2-e2). The MeCP2 gene encodes two isoforms of MeCP2 protein, referred to as MeCP2 isoform e1 and MeCP2 isoform e2, which differ in the length of their N-terminus. In some embodiments, MeCP2 isoform e1 is represented by a sequence set forth in SEQ ID NO: 1. In some embodiments, MeCP2 isoform c2 is represented by a sequence set forth in SEQ ID NO: 2.

In some embodiments, a transgene (e.g., a recombinant nucleic acid) encodes a functional fragment of MeCP2 protein (e.g., a fragment of isoform e1 or isoform e2). A “functional fragment” of MeCP2 is a truncated MeCP2 protein that retains the natural function (e.g., transcriptional activator or transcriptional repressor) of wild-type (e.g., full-length) MeCP2 protein. In some embodiments, a functional fragment of MeCP2 comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more amino acid truncations relative to full-length MeCP2 protein. In some embodiments, a functional fragment of MeCP2 comprises between about 1 and 10, 5 and 50, 20 and 100 amino acid truncations relative to full-length MeCP2 protein.

In some embodiments, a transgene (e.g., a recombinant nucleic acid) encodes a variant of MeCP2 protein (e.g., a variant of isoform e1 or isoform e2). A variant of MeCP2 protein may have between about 50% and about 99.9% identity to a wild-type MeCP2 protein (e.g., SEQ ID NO: 1 or SEQ 1D NO: 2). In some embodiments, a MeCP2 variant has about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 99% identity with a wild-type MeCP2 protein (e.g., SEQ ID NO: 1 or SEQ 1D NO: 2).

miRNA and miRNA Binding Sites

The disclosure is based, in part, on the recognition that combining miRNA regulatory elements (MREs), such as miRNA binding sites (e.g., miR-122 binding sites and miR-1 binding sites), with MREs associated with negative feedback loops regulating protein expression (e.g., miR-132 binding sites for MeCP2), simultaneously regulate transgene expression levels and de-target transgene expression in peripheral organs.

miRNAs and other small interfering nucleic acids regulate gene expression via target RNA transcript cleavage/degradation or translational repression of the target messenger RNA (mRNA), miRNAs are natively expressed, typically as final 19-25 non-translated RNA products, miRNAs exhibit their activity through sequence-specific interactions with the 3′ untranslated regions (UTR) of target mRNAs. These endogenously expressed miRNAs form hairpin precursors which are subsequently processed into a miRNA duplex, and further into a “mature” single stranded miRNA molecule. This mature miRNA guides a multiprotein complex, miRISC, which identifies target site, e.g., in the 3′ UTR regions, of target mRNAs based upon their complementarity to the mature miRNA.

The following non-limiting list of miRNA genes, and their homologues, are useful in methods and compositions of the disclosure (e.g., for mediating self-regulated expression or de-targeting of a transgene): hsa-let-7a, hsa-let-7a*, hsa-let-7b, hsa-let-7b*, hsa-let-7c, hsa-let-7c*, hsa-let-7d, hsa-let-7d*, hsa-let-7e, hsa-let-7e, hsa-let-7f, hsa-let-7f-1*, hsa-let-7f-2*, hsa-let-7g, hsa-let-7g*, hsa-let-7i, hsa-let-7i*, hsa-miR-1, hsa-miR-100, hsa-miR-100*, hsa-miR-101, hsa-miR-101*, hsa-miR-103, hsa-miR-105, hsa-miR-105*, hsa-miR-106a, hsa-miR-106a*, hsa-miR-106b, hsa-miR-106b*, hsa-miR-107, hsa-miR-10a, hsa-miR-10a*, hsa-miR-10b, hsa-miR-10b*, hsa-miR-1178, hsa-miR-1179, hsa-miR-1180, hsa-miR-1181, hsa-miR-1182, hsa-miR-1183, hsa-miR-1184, hsa-miR-1185, hsa-miR-1197, hsa-miR-1200, hsa-miR-1201, hsa-miR-1202, hsa-miR-1203, hsa-miR-1204, hsa-miR-1205, hsa-miR-1206, hsa-miR-1207-3p, hsa-miR-1207-5p, hsa-miR-1208, hsa-miR-122, hsa-miR-122*, hsa-miR-1224,3p, hsa-miR-1224-5p, hsa-miR-1225-3p, hsa-miR-1225-5p, hsa-miR-1226, hsa-miR-1226*, hsa-miR-1227, hsa-miR-1228, hsa-miR-1228*, hsa-miR-1229, hsa-miR-1231, hsa-miR-1233, hsa-miR-1234, hsa-miR-1236, hsa-miR-1237, hsa-miR-1238, hsa-miR-124, hsa-miR-124*, hsa-miR-1243, hsa-miR-1244, hsa-miR-1245, hsa-miR-1246, hsa-miR-1247, hsa-miR-1248, hsa-miR-1249, hsa-miR-1250, hsa-miR-1251, hsa-miR-1252, hsa-miR-1253, hsa-miR-1254, hsa-miR-1255a, hsa-miR-1255b, hsa-miR-1256, hsa-miR-1257, hsa-miR-1258, hsa-miR-1259, hsa-miR-125a-3p, hsa-miR-125a-5p, h % a-miR-125b, hsa-miR-125b-1*, hsa-miR-125b-2*, hsa-miR-126, hsa-miR-126*, hsa-miR-1260, hsa-miR-1261, hsa-miR-1262, hsa-miR-1263, hsa-miR-1264, hsa-miR-1265, hsa-miR-1266, hsa-miR-1267, hsa-miR-1268, hsa-miR-1269, hsa-miR-1270, hsa-miR-1271, hsa-miR-1272, hsa-miR-1273, hsa-miR-127-3p, hsa-miR-1274a, hsa-miR-1274b, hsa-miR-1275, hsa-miR-127-5p, hsa-miR-1276, hsa-miR-1277, hsa-miR-1278, hsa-miR-1279, hsa-miR-128, hsa-miR-1280, hsa-miR-1281, hsa-miR-1282, hsa-miR-1283, hsa-miR-1284, hsa-miR-1285, hsa-miR-1286, hsa-miR-1287, hsa-miR-1288, hsa-miR-1289, hsa-miR-129*, hsa-miR-1290, hsa-miR-1291, hsa-miR-1292, hsa-miR-1293, hsa-miR-129-3p, hsa-miR-1294, hsa-miR-1295, hsa-miR-129-5p, hsa-miR-1296, hsa-miR-1297, hsa-miR-1298, hsa-miR-1299, hsa-miR-1300, hsa-miR-1301, hsa-miR-1302, hsa-miR-1303, hsa-miR-1304, hsa-miR-1305, hsa-mi R-1306, hsa-miR-1307, hsa-miR-1308, hsa-miR-130a, hsa-miR-130a*, hsa-miR-130b, hsa-miR-130b*, hsa-miR-132, hsa-miR-132*, hsa-miR-1321, hsa-miR-1322, hsa-miR-1323, hsa-miR-1324, hsa-miR-133a, hsa-miR-133b, hsa-miR-134, hsa-miR-135a, hsa-miR-135a*, hsa-miR-135b, hsa-miR-135b*, hsa-miR-136, hsa-miR-136*, hsa-miR-137, hsa-miR-138, hsa-miR-138-1*, hsa-miR-138-2*, hsa-miR-139-3p, hsa-miR-139-5p, hsa-miR-140-3p, hsa-miR-140-5p, hsa-miR-141, hsa-miR-141*, hsa-miR-142-3p, hsa-miR-142-5p, hsa-miR-143, hsa-miR-143*, hsa-miR-144, hsa-miR-144*, hsa-miR-145, hsa-miR-145*, hsa-miR-146a, hsa-miR-146a*, hsa-miR-146b-3p, hsa-miR-146b-5p, hsa-miR-147, hsa-miR-147b, hsa-miR-148a, hsa-miR-148a*, hsa-miR-148b, hsa-miR-148b*, hsa-miR-149, hsa-miR-149*, hsa-miR-150, hsa-miR-150*, hsa-miR-151-3p, hsa-MiR-151-5p, hsa-miR-152, hsa-miR-153, hsa-miR-154, hsa-miR-154*, hsa-miR-155, hsa-miR-155*, hsa-miR-1Sa, hsa-miR-15a*, hsa-miR-15b, hsa-miR-15b*, hsa-miR-16, hsa-miR-16-1*, hsa-miR-16-2*, hsa-miR-17, hsa-miR-17*, hsa-miR-181a, hsa-miR-181a*, hsa-miR-181a-2*, hsa-miR-181b, hsa-miR-181c, hsa-miR-181c*, hsa-miR-181d, hsa-miR-182, hsa-miR-182*, hsa-miR-1825, hsa-miR-1826, hsa-miR-1827, hsa-miR-183, hsa-miR-183*, hsa-miR-184, hsa-miR-185, hsa-miR-185*, hsa-miR-186, hsa-miR-186*, hsa-miR-187, hsa-miR-187*, hsa-miR-188-3p, hsa-miR-188-5p, hsa-miR-18a, hsa-miR-18a*, hsa-miR-18b, hsa-miR-18b*, hsa-miR-190, hsa-miR-190b, hsa-miR-191, hsa-miR-191*, hsa-miR-192, hsa-miR-192*, hsa-miR-193a-3p, hsa-miR-193a-5p, hsa-miR-193b, hsa-miR-193b*, hsa-miR-194, hsa-miR-194*, hsa-miR-195, hsa-miR-195*, hsa-miR-196a, hsa-miR-1,96a*, hsa-miR-196b, hsa-miR-197, hsa-miR-198, hsa-miR-199a-3p, hsa-miR-199a-5p, hsa-miR-199b-5p, hsa-miR-19a, hsa-miR-19a*, hsa-miR-19b, hsa-miR-19b-1*, hsa-miR-19b-2*, hsa-miR-200a, hsa-miR-200a*, hsa-miR-200b, hsa-miR-200b*, hsa-miR-200k, hsa-miR-200c*, hsa-miR-202, hsa-miR-202*, hsa-miR-203, hsa-miR-204, hsa-miR-205, hsa-miR-206, hsa-miR-208a, hsa-miR-208b, hsa-miR-20a, hsa-miR-20a*, hsa-miR-20h, hsa-miR-20b*, hsa-miR-21, hsa-miR-21*, hsa-miR-210, hsa-miR-211, hsa-miR-212, hsa-miR-214, hsa-miR-214*, hsa-miR-215, hsa-miR-216a, hsa-miR-216b, hsa-miR-217, hsa-miR-218, hsa-miR-218-1*, hsa-miR-218-2*, hsa-miR-219-1-3p, hsa-miR-219-2-3p, hsa-miR-219-5p, hsa-miR-22, hsa-miR-22*, hsa-miR-220a, hsa-miR-220b, hsa-miR-220c, hsa-miR-221, hsa-miR-221*, hsa-miR-222, hsa-miR-222*, hsa-miR-223, hsa-miR-223*, hsa-miR-224, hsa-miR-23a, hsa-miR-23a*, hsa-miR-23b, hsa-miR-23b*, hsa-miR-24, hsa-miR-24-1*, hsa-miR-24-2*, hsa-miR-25, hsa-miR-25*, hsa-miR-26a, hsa-miR-26a-1*, hsa-miR-26a-2*, hsa-miR-26b, hsa-miR-26b*, hsa-miR-27a, hsa-miR-27a*, hsa-miR-27b, hsa-miR-27b*, hsa-miR-28-3p, hsa-miR-28-5p, hsa-miR-296-3p, hsa-miR-296-5p, hsa-miR-297, hsa-miR-298, hsa-miR-299-3p, hsa-miR-299-5p, hsa-miR-29a, hsa-miR-29a*, hsa-miR-29b, hsa-miR-29b-1*, hsa-miR-29b-2*, hsa-miR-29c, hsa-miR-29c*, hsa-miR-300, hsa-miR-301a, hsa-miR-301b, hsa-miR-302a, hsa-miR-302a*, hsa-miR-302b, hsa-miR-302b*, hsa-miR-302c, hsa-miR-302c*, hsa-miR-302d, hsa-miR-302d*, hsa-miR-302e, hsa-miR-302f, hsa-miR-30a*, hsa-miR-30a*, hsa-miR-30h, hsa-miR-30b*, hsa-miR-30c, hsa-miR-30c-1*, hsa-miR-30c-2*, hsa-miR-30d, hsa-miR-30d*, hsa-miR-30e, hsa-miR-30c*, hsa-miR-31, hsa-miR-31*, hsa-miR-32, hsa-miR-32*, hsa-miR-320a, hsa-miR-320b, hsa-miR-320c, hsa-miR-320d, hsa-miR-323-3p, hsa-miR-323-5p, hsa-miR-324-3p, hsa-miR-324-5p, hsa-miR-325, hsa-miR-326, hsa-miR-328, hsa-miR-329, hsa-miR-330-3p, hsa-miR-330-5p, hsa-miR-331-3p, hsa-miR-331-5p, hsa-miR-335, hsa-miR-335*, hsa-miR-337-3p, hsa-miR-337-5p, hsa-miR-338-3p, hsa-miR-338-5p, hsa-miR-339-3p, hsa-miR-339-5p, hsa-miR-33a, hsa-miR-33a*, hsa-miR-33b, hsa-miR-33b*, hsa-miR-340, hsa-miR-340*, hsa-miR-342-3p, hsa-miR-342-5p, hsa-miR-345, hsa-miR-346, hsa-miR-34a, hsa-miR-34a*, hsa-miR-34b, hsa-miR-34b*, hsa-miR-34c-3p, hsa-miR-34c-5p, hsa-miR-361-3p, hsa-miR-361-5p, hsa-miR-362-3p, hsa-miR-362-5p, hsa-miR-363, hsa-miR-363*, hsa-miR-365, hsa-miR-367, hsa-miR-367*, hsa-miR-369-3p, hsa-miR-369-5p, hsa-miR-370, hsa-miR-371-3p, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373, hsa-miR-373*, hsa-miR-374a, hsa-miR-374a*, hsa-miR-374b, hsa-miR-374b*, hsa-miR-375, hsa-miR-376a, hsa-miR-376a*-hsa-miR-376b, hsa-miR-376c, hsa-miR-377, hsa-miR-377*, hsa-miR-378, hsa-miR-378*, hsa-miR-379, hsa-miR-379*, hsa-miR-380, hsa-miR-380*, hsa-miR-381, hsa-miR-382, hsa-miR-383, hsa-miR-384, hsa-miR-409-3p, hsa-miR-409-5p, hsa-miR-410, hsa-miR-411, hsa-miR-411*, hsa-miR-412, hsa-miR-421, hsa-miR-422a, hsa-miR-423-3p, hsa-miR-423-5p, hsa-miR-424, hsa-miR-424*, hsa-miR-425, hsa-miR-425*, hsa-miR-429, hsa-miR-431, hsa-miR-431*, hsa-miR-432, hsa-miR-432*, hsa-miR-433, hsa-miR-448, hsa-miR-449a, hsa-miR-449b, hsa-miR-450a, hsa-miR-450b-3p, hsa-miR-450h-5p, hsa-miR-451, hsa-miR-452, hsa-miR-452*, hsa-miR-453, hsa-miR-454, hsa-miR-454*, hsa-miR-455-3p, hsa-miR-455-5p, hsa-miR-483-3p, hsa-miR-483-5p, hsa-miR-484, hsa-miR-485-3p, hsa-miR-485-5p, hsa-miR-486-3p, hsa-miR-486-5p, h-a-miR-487u, hsa-miR-487b, ha-miR-488, hsa-miR-488*, hsa-miR-489, h-a-miR-490-3p, hsa-miR-490-5p, hsa-miR-491-3p, hsa-miR-491-5p, hsa-miR-492, hsa-miR-493, hsa-miR-493*, hsa-miR-494, hsa-miR-495, hsa-miR-496, hsa-miR-497, hsa-miR-497*, hsa-miR-498, hsa-miR-499-3p, hsa-miR-499-5p, hsa-miR-500, hsa-miR-500*, hsa-miR-501-3p, hsa-miR-501-5p, hsa-miR-502-3p, hsa-miR-502-5p, hsa-miR-503, hsa-miR-504, hsa-miR-505, hsa-miR-505*, hsa-miR-506, hsa-miR-507, hsa-miR-508-3p, hsa-miR-508-5p, hsa-miR-509-3-5p, hsa-miR-509-3p, hsa-miR-509-5p, hsa-miR-510, hsa-miR-511, hsa-miR-512-3p, hsa-miR-512-5p, hsa-miR-513a-3p, hsa-miR-513u-5p, hsa-miR-5I3b, hsa-miR-513c, hsa-miR-514, hsa-miR-515-3p, hsa-miR-515-5p, hsa-miR-516a-3p, hsa-miR-516a-5p, hsa-miR-516b, hsa-miR-517*, hsa-miR-517a, hsa-miR-517b, hsa-miR-517c, hsa-miR-518a-3p, hsa-miR-518a-5p, hsa-miR-518b, hsa-miR-518c, hsa-miR-518e*, hsa-miR-518d-3p, hsa-miR-518d-5p, hsa-miR-518e, hsa-miR-518c*, hsa-miR-518f, hsa-miR-518f*, hsa-miR-519a, hsa-miR-519b-3p, hsa-miR-519c-3p, hsa-miR-519d, hsa-miR-519e, hsa-miR-519e*, hsa-miR-520a-3p, hsa-miR-520a-5p, hsa-miR-520b, hsa-miR-520c-3p, hsa-miR-520d-3p, hsa-miR-520d-5p, hsa-miR-520e, hsa-miR-520f, hsa-miR-520g, hsa-miR-520h, hsa-miR-521, hsa-miR-522, hsa-miR-523, hsa-miR-524-3p, hsa-miR-524-5p, hsa-miR-525-3p, hsa-miR-525-5p, hsa-miR-526b, hsa-miR-526b*, hsa-miR-532-3p, hsa-miR-532-5p, hsa-miR-539, hsa-miR-541, hsa-miR-541*, hsa-miR-542-3p, hsa-miR-542-5p, hsa-miR-543, hsa-miR-544, hsa-miR-545, hsa-miR-545*, hsa-miR-548a-3p, hsa-miR-548a-5p, hsa-miR-548b-3p, hsa-miR-548b-5p, hsa-miR-548c-3p, hsa-miR-548c-5p, hsa-miR-548d-3p, hsa-miR-548d-5p, hsa-miR-548c, hsa-miR-548f, hsa-miR-548g, hsa-miR-548h, hsa-miR-548i, hsa-miR-548j, hsa-miR-548k, hsa-miR-548l, hsa-miR-548m, hsa-miR-548n, hsa-miR-548o, hsa-miR-548p, hsa-miR-549, hsa-miR-550, hsa-miR-550*, hsa-miR-551a, hsa-miR-551b, hsa-miR-551b*, hsa-miR-552, hsa-miR-553, hsa-miR-554, hsa-miR-555, hsa-miR-556-3p, hsa-miR-556-5p, hsa-miR-557, hsa-miR-558, hsa-miR-559, hsa-miR-561, hsa-miR-562, hsa-miR-563, hsa-miR-564, hsa-miR-566, hsa-miR-567, hsa-miR-568, hsa-miR-569, hsa-miR-570, hsa-miR-571, hsa-miR-572, hsa-miR-573, hsa-miR-574-3p, hsa-miR-574-5p, hsa-miR-575, hsa-miR-576-3p, hsa-miR-576-5p, hsa-miR-577, hsa-miR-578, hsa-miR-579, hsa-miR-580, hsa-miR-581, hsa-miR-582-3p, hsa-miR-582-5p, hsa-miR-583, hsa-miR-584, hsa-miR-585, hsa-miR-586, hsa-miR-587, hsa-miR-588, hsa-miR-589, hsa-miR-589*, hsa-miR-590-3p, hsa-miR-590-5p, hsa-miR-591, hsa-miR-592, hsa-miR-593, hsa-miR-593*, hsa-miR-595, hsa-miR-596, hsa-miR-597, hsa-miR-598, hsa-miR-599, hsa-miR-600, hsa-miR-601, hsa-miR-602, hsa-miR-603, hsa-miR-604, hsa-miR-605, hsa-miR-606, hsa-miR-607, hsa-miR-608, hsa-miR-609, hsa-miR-610, hsa-miR-611, hsa-miR-612, hsa-miR-613, hsa-miR-614, hsa-miR-615-3p, hsa-miR-615-5p, hsa-miR-616, hsa-miR-616*, hsa-miR-617, hsa-miR-618, hsa-miR-619, hsa-miR-620, hsa-miR-62I, hsa-miR-622, hsa-miR-623, hsa-miR-624, hsa-miR-624*, hsa-miR-625, hsa-miR-625*, hsa-miR-626, hsa-miR-627, hsa-miR-628-3p, hsa-miR-628-5p, hsa-miR-629, hsa-miR-629*, hsa-miR-630, hsa-miR-631, hsa-miR-632, hsa-miR-633, hsa-miR-634, hsa-miR-635, hsa-miR-636, hsa-miR-637, hsa-miR-638, hsa-miR-639, hsa-miR-640, hsa-miR-641, hsa-miR-642, hsa-miR-643, hsa-miR-644, hsa-miR-645, hsa-miR-646, hsa-miR-647, hsa-miR-648, hsa-miR-649, hsa-miR-650, h % a-miR-651, hsa-miR-652, hsa-miR-653, hsa-miR-654-3p, hsa-miR-654-5p, hsa-miR-655, hsa-miR-656, hsa-miR-657, hsa-miR-658, hsa-miR-659, hsa-miR-660, hsa-miR-661, hsa-miR-662, hsa-miR-663, hsa-miR-663b, hsa-miR-664, hsa-miR-664*, hsa-miR-665, hsa-miR-668, hsa-miR-671-3p, hsa-miR-671-5p, hsa-miR-675, hsa-miR-7, hsa-miR-708, hsa-miR-708*, hsa-miR-7-1*, hsa-miR-7-2′*, hsa-miR-720, hsa-miR-744, hsa-miR-744*, hsa-miR-758, hsa-miR-760, hsa-miR-765, hsa-miR-766, hsa-miR-767-3p, hsa-miR-767-5p, hsa-miR-7 68-3p, hsa-miR-768-5p, hsa-miR-769-3p, hsa-miR-769-5p, hsa-miR-770-5p, hsa-miR-802, hsa-miR-873, hsa-miR-874, hsa-miR-875-3p, hsa-miR-875-5p, hsa-miR-876-3p, hsa-miR-876-5p, hsa-miR-877, hsa-miR-877*, hsa-miR-885-3p, hsa-miR-885-5p, hsa-miR-886-3p, hsa-miR-886-5p, hsa-miR-887, hsa-miR-888, hsa-miR-888*, hsa-miR-889, hsa-miR-890, hsa-miR-891a, hsa-miR-891 b, hsa-miR-892a, hsa-miR-892b, hsa-miR-9, hsa-miR-9*, hsa-miR-920, hsa-miR-921, hsa-miR-922, hsa-miR-923, hsa-miR-924, hsa-miR-92a, hsa-miR-92a-1*, hsa-miR-92a-2*, hsa-miR-92b, hsa-miR-92b*, hsa-miR-93, hsa-miR-93*, hsa-miR-933, hsa-miR-934, hsa-miR-935, hsa-miR-936, hsa-miR-937, hsa-miR-938, hsa-miR-939, hsa-miR-940, hsa-miR-941, hsa-miR-942, hsa-miR-943, hsa-miR-944, hsa-miR-95, hsa-miR-96, hsa-miR-96*, hsa-miR-98, hsa-miR-99a, hsa-miR-99a*, hsa-miR-99b, and hsa-miR-99b*.

In some embodiments, one or more binding sites of a construct as described by the disclosure (e.g., recombinant nucleic acid, AAV vector, rAAV, etc.) de-targets transgene expression from a cell of the immune system (e.g., an antigen presenting cell (APC)). In some embodiments, an miRNA that de-targets transgene expression from an immune cell (e.g., an antigen presenting cell) is referred to as an immune-associated miRNA. In some embodiments, an immune-associated miRNA is an miRNA expressed in immune cells that exhibits at least a 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold higher level of expression in an immune cell compared with a non-immune cell (e.g., a control cell, such as a HeLa cell, HEK293 cell, mesenchymal cell, etc.). In some embodiments, the cell of the immune system (immune cell) in which the immune-associated miRNA is expressed is a B cell, T cell, Killer T cell, Helper T cell, γδ T cell, dendritic cell, macrophage, monocyte, vascular endothelial cell, or other immune cell. In some embodiments, the cell of the immune system is a B cell expressing one or more of the following markers: B220, BLAST-2 (EBVCS), Bu-1, CD19, CD20 (26), CD22, CD24, CD27, CD57, CD72, CD79a, CD79b, CD86, chB6, D8/17, FMC7, L26, M17, MUM-1, Pax-5 (BSAP), and PC47H. In some embodiments, the cell of the immune system is a T cell expressing one or more of the following markers: ART2, CD1a, CD1d, CD11b (Mae-1), CD134 (OX40), CD150, CD2, CD25 (interleukin 2 receptor alpha), CD3, CD38, CD4, CD45RO, CD5, CD7, CD72, CD8, CRTAM, FOXP3, FT2, GPCA, HLA-DR, HML-1, HT23A, Leu-22, Ly-2, Ly-m22, MICG, MRC OX 8, MRC OX-22, OX40, PD-1 (Programmed death-1), RT6, TCR (T cell receptor), Thy-1 (CD90), and TSA-2 (Thymic shared Ag-2). In some embodiments, an immune-associated miRNA is selected from: miR-15a, miR-16-1, miR-17, miR-18a, miR-19a, miR-19b-1, miR-20a, miR-21, miR-29a/b/c, miR-30b, miR-31, miR-34a, miR-92a-1, miR-106a, miR-125a/b, miR-142-3p, miR-146a, miR-150, miR-155, miR-181a, miR-223 and miR-424, miR-221, miR-222, let-7i, miR-148, and miR-152.

Recombinant AAV Vectors (rAAV Vectors)

“Recombinant AAV (rAAV) vectors” of the disclosure are typically composed of, at a minimum, a transgene and its regulatory sequences, and 5′ and 3′ AAV inverted terminal repeats (ITRs). It is this recombinant AAV vector which is packaged into a capsid protein and delivered to a selected target cell. In some embodiments, the transgene is a nucleic acid sequence, heterologous to the vector sequences, which encodes a polypeptide, protein, functional RNA molecule (e.g., gRNA) or other gene product, of interest. The nucleic acid coding sequence is operatively linked to regulatory components in a manner which permits transgene transcription, translation, and/or expression in a cell of a target tissue.

In some embodiments, the instant disclosure relates to a recombinant AAV (rAAV) vector comprising a nucleic acid sequence including a promoter operably linked to a transgene, wherein the transgene encodes a MeCP2 protein (e.g., MeCP2 isoform e1). In some embodiments, a rAAV vector further comprises nucleic acid sequences encoding one or more AAV inverted terminal repeat sequences (ITRs), for example AAV2 ITRs. In some embodiments, a rAAV vector further comprises nucleic acid sequences encoding one or more AAV ITRs selected from the group consisting of AAV3, AAV4, AAV5, and AAV6. In some embodiments, ITRs are artificial sequences that replace ITR function, for example as disclosed in WO/2016/172008.

The AAV sequences of the vector typically comprise the cis-acting 5′ and 3′ inverted terminal repeat sequences (See, e.g., B. J. Carter, in “Handbook of Parvoviruses”, ed., P. Tijsser. CRC Press, pp. 155 168 (1990)). The ITR sequences are about 145 bp in length. Preferably, substantially the entire sequences encoding the ITRs are used in the molecule, although some degree of minor modification of these sequences is permissible. The ability to modify these ITR sequences is within the skill of the art. (See, e.g., texts such as Sambrook et al. “Molecular Cloning. A Laboratory Manual”, 2d ed., Cold Spring Harbor Laboratory, New York (1989); and K. Fisher et al., J Virol., 70:520 532 (1996)). An example of such a molecule employed in the present disclosure is a “cis-acting” plasmid containing the transgene, in which the selected transgene sequence and associated regulatory elements are flanked by the 5′ and 3′ AAV ITR sequences. The AAV ITR sequences may be obtained from any known AAV, including presently identified mammalian AAV types (e.g., AAV2. AAV3, AAV4. AAV5, or AAV6 ITR sequences).

In addition to the major elements identified above for the recombinant AAV vector, the vector also includes control elements necessary which are operably linked to the transgene in a manner which permits its transcription, translation and/or expression in a cell transfected with the plasmid vector or infected with the virus produced by the disclosure. As used herein, “operably linked” sequences include both expression control sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.

Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation (polyA) signals; sequences that stabilize cytoplasmic mRNA: sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance secretion of the encoded product. A great number of expression control sequences, including promoters which are native, constitutive, inducible and/or tissue-specific, are known in the art and may be utilized.

As used herein, a nucleic acid sequence (e.g., coding sequence) and regulatory sequences are said to be “operably” linked when they are covalently linked in such a way as to place the expression or transcription of the nucleic acid sequence under the influence or control of the regulatory sequences. If it is desired that the nucleic acid sequences be translated into a functional protein, two DNA sequences are said to be operably linked if induction of a promoter in the 5′ regulatory sequences results in the transcription of the coding sequence and if the nature of the linkage between the two DNA sequences does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the promoter region to direct the transcription of the coding sequences, or (3) interfere with the ability of the corresponding RNA transcript to be translated into a protein. Thus, a promoter region would be operably linked to a nucleic acid sequence if the promoter region were capable of effecting transcription of that DNA sequence such that the resulting transcript might be translated into the desired protein or polypeptide. Similarly two or more coding regions are operably linked when they are linked in such a way that their transcription from a common promoter results in the expression of two or more proteins having been translated in frame. In some embodiments, operably linked coding sequences yield a fusion protein. In some embodiments, operably linked coding sequences yield a functional RNA (e.g., gRNA, miRNA).

For nucleic acids encoding proteins, a polyadenylation sequence generally is inserted following the transgene sequences and before the 3′ AAV ITR sequence. A rAAV construct useful in the present disclosure may also contain an intron, desirably located between the promoter/enhancer sequence and the transgene. One possible intron sequence is derived from SV40, and is referred to as the SV-40 T intron sequence. Another vector element that may be used is an internal ribosome entry site (IRES). An IRES sequence is used to produce more than one polypeptide from a single gene transcript. An IRES sequence would be used to produce a protein that contain more than one polypeptide chains. Selection of these and/or other vector elements may be performed, as appropriate, and many such sequences are available [see, e.g., Sambrook et al. and references cited therein at, for example, pages 3.18 3.26 and 16.17 16.27 and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1989]. In some embodiments, a Foot and Mouth Disease Virus 2A sequence is included in polyprotein: this is a small peptide (approximately 18 amino acids in length) that has been shown to mediate the cleavage of polyproteins (Ryan, M D et aL, EMBO, 1994; 4: 928-933; Mattion, N M et al., J Virology, November 1996; p. 8124-8127; Furler, S et al., Gene Therapy, 2001: 8: 864-873: and Halpin. C et al., The Plant Journal, 1999; 4: 453-459). The cleavage activity of the 2A sequence has previously been demonstrated in artificial systems including plasmids and gene therapy vectors (AAV and retroviruses) (Ryan. M D et al., EMBO, 1994; 4: 928-933; Mattion. N M et al., J Virology, November 1996; p. 8124-8127; Furler, S et al., Gene Therapy, 2001: 8: 864-873: and Halpin, C et al., The Plant Journal, 1999; 4: 453-459; de Felipe, P et al., Gene Therapy, 1999; 6: 198-208: de Felipe, P et al., Human Gene Therapy, 2000: 11: 1921-1931.; and Klump, H et al., Gene Therapy, 2001: 8: 811-817).

The precise nature of the regulatory sequences needed for gene expression in host cells may vary between species, tissues or cell types, but shall in general include, as necessary, 5′ non-transcribed and 5′ non-translated sequences involved with the initiation of transcription and translation respectively, such as a TATA box, capping sequence, CAAT sequence, enhancer elements, and the like. Especially, such 5′ non-transcribed regulatory sequences will include a promoter region that includes a promoter sequence for transcriptional control of the operably joined gene. Regulatory sequences may also include enhancer sequences or upstream activator sequences as desired. The vectors of the disclosure may optionally include 5′ leader or signal sequences. The choice and design of an appropriate vector is within the ability and discretion of one of ordinary skill in the art.

Examples of constitutive promoters include, without limitation, the retroviral Rous sarcoma virus (RSV) LTR promoter (optionally with the RSV enhancer), the cytomegalovirus (CMV) promoter (optionally with the CMV enhancer) [see. e.g., Boshart et al, Cell, 41:521-530 (1985)], the SV40 promoter, the dihydrofolate reductase promoter, the β-actin promoter, the phosphoglycerol kinase (PGK) promoter, and the EF1α promoter [Invitrogen]. In some embodiments, a promoter is an enhanced chicken β-actin promoter.

Inducible promoters allow regulation of gene expression and can be regulated by exogenously supplied compounds, environmental factors such as temperature, or the presence of a specific physiological state, e.g., acute phase, a particular differentiation state of the cell, or in replicating cells only. Inducible promoters and inducible systems are available from a variety of commercial sources, including, without limitation, Invitrogen, Clontech and Ariad. Many other systems have been described and can be readily selected by one of skill in the art. Examples of inducible promoters regulated by exogenously supplied promoters include the zinc-inducible sheep metallothionine (MT) promoter, the dexamethasone (Dex)-inducible mouse mammary tumor virus (MMTV) promoter, the T7 polymerase promoter system (WO 98/10088); the ecdysone insect promoter (No et al, Proc. Natl. Acad. Sci. USA, 93:3346-3351 (1996)), the tetracycline-repressible system (Gossen et al, Proc. Natl. Acad. Sci. USA, 89:5547-5551 (1992)), the tetracycline-inducible system (Gossen et al. Science, 268:1766-1769 (1995), see also Harvey et al, Curr. Opin. Chem. Biol., 2:512-518 (1998)), the RU486-inducible system (Wang et al, Nat. Biotech., 15:239-243 (1997) and Wang et al, Gene Ther., 4:432-441 (1997)) and the rapamycin-inducible system (Magari et al, J. Clin. Invest., 100:2865-2872 (1997)). Still other types of inducible promoters which may be useful in this context are those which are regulated by a specific physiological state, e.g., temperature, acute phase, a particular differentiation state of the cell, or in replicating cells only.

In another embodiment, the native promoter for the transgene will be used. The native promoter may be preferred when it is desired that expression of the transgene should mimic the native expression. The native promoter may be used when expression of the transgene must be regulated temporally or developmentally, or in a tissue-specific manner, or in response to specific transcriptional stimuli. For example, in some embodiments, a native promoter is a MeCP2 promoter, such as a mouse MeCP2 promoter. In some embodiments, a mouse MeCP2 promoter is represented by a sequence set forth in SEQ ID NO: 3. In a further embodiment, other native expression control elements, such as enhancer elements, polyadenylation sites or Kozak consensus sequences may also be used to mimic the native expression.

In some embodiments, the regulatory sequences impart tissue-specific gene expression capabilities. In some cases, the tissue-specific regulatory sequences bind tissue-specific transcription factors, that induce transcription in a tissue specific manner. Such tissue-specific regulatory sequences (e.g., promoters, enhancers, etc.,) are well known in the art. Exemplary tissue-specific regulatory sequences include, but are not limited to the following tissue specific promoters: an eye-specific retinoschisin promoter or K12 promoter, a liver-specific thyroxin binding globulin (TBG) promoter, an insulin promoter, a glucagon promoter, a somatostatin promoter, a pancreatic polypeptide (PPY) promoter, a synapsin-1 (Syn) promoter, a creatine kinase (MCK) promoter, a mammalian desmin (DES) promoter, a α-myosin heavy chain (a-MHC) promoter, or a cardiac Troponin T (cTnT) promoter. Other exemplary promoters include Beta-actin promoter, hepatitis B virus core promoter. Sandig et al., Gene Ther., 3:1002-9 (1996); alpha-fetoprotein (AFP) promoter, Arbuthnot et al., Hum. Gene Ther., 7:1503-14 (1996)), bone osteocalcin promoter (Stein et al., Mol. Biol. Rep., 24:185-96 (1997)); bone sialoprotein promoter (Chen et al., J. Bone Miner. Res., 11:654-64 (1996)), CD2 promoter (Hansal et al., J. Immunol., 161:1063-8 (1998); immunoglobulin heavy chain promoter: T cell receptor α-chain promoter, neuronal such as neuron-specific enolase (NSE) promoter (Andersen et at, Cell. Mol. Neurobiol., 13:503-15 (1993)), neurofilament light-chain gene promoter (Piccioli et al., Proc. Natl. Acad. Sci. USA, 88:5611-5 (1991)), and the neuron-specific vgf gene promoter (Piccioli et al., Neuron, 15:373-84 (1995)), among others which will be apparent to the skilled artisan.

In some embodiments, one or more bindings sites for one or more of miRNAs are incorporated in a transgene of a rAAV vector, to inhibit the expression of the transgene in one or more tissues of an subject harboring the transgene. The skilled artisan will appreciate that binding sites may be selected to control the expression of a transgene in a tissue specific manner. For example, binding sites for the liver-specific miR-122 may be incorporated into a transgene to inhibit expression of that transgene in the liver. The target sites in the mRNA may be in the 5′ UTR, the 3 UTR or in the coding region. Typically, the target site is in the 3′ UTR of the mRNA. Furthermore, the transgene may be designed such that multiple miRNAs regulate the mRNA by recognizing the same or multiple sites. The presence of multiple miRNA binding sites may result in the cooperative action of multiple RISCs and provide highly efficient inhibition of expression. The target site sequence may comprise a total of 5-10), 10-60, or more nucleotides. The target site sequence may comprise at least 5 nucleotides of the sequence of a target gene binding site.

Recombinant Adeno-Associated Viruses (rAAVs)

In some aspects, the disclosure provides isolated AAVs. As used herein with respect to AAVs, the term “isolated” refers to an AAV that has been artificially produced or obtained. Isolated AAVs may be produced using recombinant methods. Such AAVs are referred to herein as “recombinant AAVs”. Recombinant AAVs (rAAVs) preferably have tissue-specific targeting capabilities, such that a nuclease and/or transgene of the rAAV will be delivered specifically to one or more predetermined tissue(s). The AAV capsid is an important element in determining these tissue-specific targeting capabilities. Thus, an rAAV having a capsid appropriate for the tissue being targeted can be selected.

Methods for obtaining recombinant AAVs having a desired capsid protein are well known in the art. (See, for example, US 2003/0138772), the contents of which are incorporated herein by reference in their entirety). Typically the methods involve culturing a host cell which contains a nucleic acid sequence encoding an AAV capsid protein; a functional rep gene; a recombinant AAV vector composed of, AAV inverted terminal repeats (ITRs) and a transgene; and sufficient helper functions to permit packaging of the recombinant AAV vector into the AAV capsid proteins. In some embodiments, capsid proteins are structural proteins encoded by the cap gene of an AAV. AAVs comprise three capsid proteins, virion proteins 1 to 3 (named VP1, VP2 and VP3), all of which are transcribed from a single cap gene via alternative splicing. In some embodiments, the molecular weights of VP1, VP2 and VP3 are respectively about 87 kDa, about 72 kDa and about 62 kDa. In some embodiments, upon translation, capsid proteins form a spherical 60-mer protein shell around the viral genome. In some embodiments, the functions of the capsid proteins are to protect the viral genome, deliver the genome and interact with the host. In some aspects, capsid proteins deliver the viral genome to a host in a tissue specific manner.

In some embodiments, an rAAV described by the disclosure comprises one or mom capsid proteins capable of crossing the blood-brain barrier. In some embodiments, the at least one capsid protein has a serotype selected from the group consisting of AAV1, AAV2, AAV2i8, AAV2.5, AAV6, AAV8, AAVrh8, AAV9, AAVrh10, AAV-B1, AAV9.45A-String (e.g., AAV9.45-AS), AAV9.45Angiopep, AAV9.47-Angiopep, and AAV9.47-AS. In some embodiments, the at least one capsid protein has a AAV-PHP.B serotype, for example as described in U.S. Pat. No. 9,585,971. In some embodiments, a capsid protein has a serotype as described in WO2015/127128, WO2016/054554, WO2016/054557, or WO2016/065001.

The components to be cultured in the host cell to package a rAAV vector in an AAV capsid may be provided to the host cell in trans. Alternatively, any one or more of the required components (e.g., recombinant AAV vector, rep sequences, cap sequences, and/or helper functions) may be provided by a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art. Most suitably, such a stable host cell will contain the required component(s) under the control of an inducible promoter. However, the required component(s) may be under the control of a constitutive promoter. Examples of suitable inducible and constitutive promoters are provided herein, in the discussion of regulatory elements suitable for use with the transgene. In still another alternative, a selected stable host cell may contain selected component(s) under the control of a constitutive promoter and other selected component(s) under the control of one or more inducible promoters. For example, a stable host cell may be generated which is derived from 293 cells (which contain E1 helper functions under the control of a constitutive promoter), but which contain the rep and/or cap proteins under the control of inducible promoters. Still other stable host cells may be generated by one of skill in the art.

In some embodiments, the instant disclosure relates to a host cell containing a nucleic acid that comprises a coding sequence encoding a protein (e.g., MeCP2 protein, such as MeCP2 isoform e1). In some embodiments, the instant disclosure relates to a composition comprising the host cell described above. In some embodiments, the composition comprising the host cell above further comprises a cryopreservative.

The recombinant AAV vector, rep sequences, cap sequences, and helper functions required for producing the rAAV of the disclosure may be delivered to the packaging host cell using any appropriate genetic element (vector). The selected genetic element may be delivered by any suitable method, including those described herein. The methods used to construct any embodiment of this disclosure are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See. e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limitation on the present disclosure. See. e.g., K. Fisher et al., J. Virol., 70:520-532 (1993) and U.S. Pat. No. 5,478,745.

In some embodiments, recombinant AAVs may be produced using the triple transfection method (described in detail in U.S. Pat. No. 6,001,650). Typically, the recombinant AAVs are produced by transfecting a host cell with an recombinant AAV vector (comprising a transgene) to be packaged into AAV particles, an AAV helper function vector, and an accessory function vector. An AAV helper function vector encodes the “AAV helper function” sequences (i.e., rep and cap), which function in trans for productive AAV replication and encapsidation. Preferably, the AAV helper function vector supports efficient AAV vector production without generating any detectable wild-type AAV virions (i.e., AAV virions containing functional rep and cap genes). Non-limiting examples of vectors suitable for use with the present disclosure include pHLP19, described in U.S. Pat. No. 6,001,650 and pRep6cap6 vector, described in U.S. Pat. No. 6,156,303, the entirety of both incorporated by reference herein. The accessory function vector encodes nucleotide sequences for non-AAV derived viral and/or cellular functions upon which AAV is dependent for replication (i.e., “accessory functions”). The accessory functions include those functions required for AAV replication, including, without limitation, those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of cap expression products, and AAV capsid assembly. Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1), and vaccinia virus.

In some aspects, the disclosure provides transfected host cells. The term “transfection” is used to refer to the uptake of foreign DNA by a cell, and a cell has been “transfected” when exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques are generally known in the art. See, e.g., Giaham et al. (1973) Virology, 52:456, Sambrook et al. (1989) Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York. Davis et al. (1986) Basic Methods in Molecular Biology, Elsevier. and Chu et al. (1981) Gene 13:197. Such techniques can be used to introduce one or more exogenous nucleic acids, such as a nucleotide integration vector and other nucleic acid molecules, into suitable host cells.

A “host cell” refers to any cell that harbors, or is capable of harboring, a substance of interest. Often a host cell is a mammalian cell. A host cell may be used as a recipient of an AAV helper construct, an AAV minigene plasmid, an accessory function vector, or other transfer DNA associated with the production of recombinant AAVs. The term includes the progeny of the original cell which has been transfected. Thus, a “host cell” as used herein may refer to a cell which has been transfected with an exogenous DNA sequence. It is understood that the progeny of a single parental cell may not necessarily be completely identical in morphology or in genomic or total DNA complement as the original parent, due to natural, accidental, or deliberate mutation.

As used herein, the term “cell line” refers to a population of cells capable of continuous or prolonged growth and division in vitro. Often, cell lines are clonal populations derived from a single progenitor cell. It is further known in the art that spontaneous or induced changes can occur in karyotype during storage or transfer of such clonal populations. Therefore, cells derived from the cell line referred to may not be precisely identical to the ancestral cells or cultures, and the cell line referred to includes such variants.

As used herein, the terms “recombinant cell” refers to a cell into which an exogenous DNA segment, such as DNA segment that leads to the transcription of a biologically-active polypeptide or production of a biologically active nucleic acid such as an RNA, has been introduced.

As used herein, the term “vector” includes any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, artificial chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors. In some embodiments, useful vectors are contemplated to be those vectors in which the nucleic acid segment to be transcribed is positioned under the transcriptional control of a promoter. A “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. The phrases “operatively positioned,” “under control” or “under transcriptional control” means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene. The term “expression vector or construct” means any type of genetic construct containing a nucleic acid in which part or all of the nucleic acid encoding sequence is capable of being transcribed. In some embodiments, expression includes transcription of the nucleic acid, for example, to generate a biologically-active polypeptide product or functional RNA (e.g., guide RNA) from a transcribed gene. The foregoing methods for packaging recombinant vectors in desired AAV capsids to produce the rAAVs of the disclosure are not meant to be limiting and other suitable methods will be apparent to the skilled artisan.

Administration Methods

Compositions described by the disclosure (e.g., recombinant nucleic acids, rAAVs, pharmaceutical compositions, etc.) may be delivered to a subject according to any appropriate methods known in the art. Compositions (e.g., recombinant nucleic acids, rAAVs, pharmaceutical compositions, etc.), preferably suspended in a physiologically compatible carrier (i.e., in a composition), may be administered to a subject, i.e. host animal, such as a human, mouse, rat, cat, dog, sheep, rabbit, horse, cow, goat, pig, guinea pig, hamster, chicken, turkey, or a non-human primate (e.g., Macaque). In some embodiments, a host animal does not include a human. In some embodiments, a subject is a human. In some embodiments, a subject is less than a year old, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 months old.

Delivery of the compositions to a mammalian subject may be by, for example, systemic injection (e.g., intravenous injection) or intrathecal injection. Additional methods of administering compositions to the CNS of a subject, for example intracranial injection, intrastriatal injection, etc. may also be used. Combinations of administration methods (e.g., topical administration and intrastromal injection) can also be used.

In some embodiments, the compositions of the disclosure may comprise an rAAV alone, or in combination with one or more other viruses (e.g., a second rAAV encoding having one or more different transgenes). In some embodiments, a composition comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more different rAAVs each having one or more different transgenes.

In some embodiments, a composition further comprises a pharmaceutically acceptable carrier. Suitable carriers may be readily selected by one of skill in the art in view of the indication for which the composition is directed. For example, one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (e.g., phosphate buffered saline). Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water. The selection of the carrier is not a limitation of the present disclosure.

Optionally, the compositions of the disclosure may contain, in addition to the recombinant nucleic acid or rAAV and carrier(s), other pharmaceutical ingredients, such as preservatives, or chemical stabilizers. Suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and parachlorophenol. Suitable chemical stabilizers include gelatin and albumin.

The compositions are administered in sufficient amounts to transfect the cells of a desired tissue (e.g., CNS tissue) and to provide sufficient levels of gene transfer and expression without undue adverse effects. Examples of pharmaceutically acceptable mutes of administration include, but am not limited to, direct delivery to the selected organ (e.g., intrastromal delivery to the eye), oral, inhalation (including intranasal and intratracheal delivery), intraocular, intravenous, intramuscular, subcutaneous, intradermal, intratumoral, and other parental routes of administration. Routes of administration may be combined, if desired.

The dose of rAAV virions required to achieve a particular “therapeutic effect,” e.g., the units of dose in genome copies/per kilogram of body weight (GC/kg), will vary based on several factors including, but not limited to: the route of rAAV virion administration, the level of gene or RNA expression required to achieve a therapeutic effect, the specific disease or disorder being treated, and the stability of the gene or RNA product. One of skill in the art can readily determine a rAAV virion dose range to treat a patient having a particular disease or disorder based on the aforementioned factors, as well as other factor.

An effective amount of composition (e.g., recombinant nucleic acid, rAAV, pharmaceutical composition, etc.) is an amount sufficient to target infect an animal, target a desired tissue. In some embodiments, an effective amount of an rAAV is an amount sufficient to produce a stable somatic transgenic animal model. The effective amount will depend primarily on factors such as the species, age, weight, health of the subject, and the tissue to be targeted, and may thus vary among animal and tissue. For example, an effective amount of the rAAV is generally in the range of from about 1 ml to about 100 ml of solution containing from about 109 to 1016 genome copies. In some cases, a dosage between about 1011 to 1013 rAAV genome copies is appropriate. In certain embodiments, 1010 or 1011 rAAV genome copies is effective to target CNS tissue (e.g., corneal tissue). In some cases, stable transgenic animals are produced by multiple doses of an rAAV.

In some embodiments, a dose of the composition is administered to a subject no more than once per calendar day (e.g., a 24-hour period). In some embodiments, a dose of the composition is administered to a subject no more than once per 3, 4, 5, 6, or 7 calendar days. In some embodiments, a dose of the composition is administered to a subject no more than once per calendar week (e.g., 7 calendar days). In some embodiments, a dose of the composition is administered to a subject no more than bi-weekly (e.g., once in a two calendar week period). In some embodiments, a dose of the composition is administered to a subject no more than once per calendar month (e.g., once in 30 calendar days). In some embodiments, a dose of the composition is administered to a subject no more than once per six calendar months. In some embodiments, a dose of the composition is administered to a subject no more than once per calendar year (e.g., 365 days or 366 days in a leap year).

In some embodiments, compositions are formulated to reduce aggregation of AAV particles in the composition, particularly where high rAAV concentrations are present (e.g., ˜1013 GC/ml or more). Appropriate methods for reducing aggregation of may be used, including, for example, addition of surfactants, pH adjustment, salt concentration adjustment, etc. (See, e.g., Wright F R, et al., Molecular Therapy (2005) 12, 171-178, the contents of which are incorporated herein by reference.)

Formulation of pharmaceutically-acceptable excipients and carrier solutions is well-known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens. Typically, these formulations may contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 70% or 80% or more of the weight or volume of the total formulation. Naturally, the amount of active compound in each therapeutically-useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.

In some embodiments, compositions (e.g., recombinant nucleic acids, rAAVs, pharmaceutical compositions, etc.) in suitably formulated pharmaceutical compositions disclosed herein are delivered directly to target tissue, e.g., direct to CNS tissue (e.g., brain, spinal cord, etc.) However, in certain circumstances it may be desirable to separately or in addition deliver the compositions via another route, e.g., subcutaneously, intraopancreatically, intranasally, parenterally, intravenously, intramuscularly, intrathecally, or orally, intraperitoneally, or by inhalation. In some embodiments, the administration modalities as described in U.S. Pat. Nos. 5,543,158; 5,641,515 and 5,399,363 (each specifically incorporated herein by reference in its entirety) may be used to deliver rAAVs. In some embodiments, a preferred mode of administration is by intravenous injection.

The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. In many cases the form is sterile and fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

For administration of an injectable aqueous solution, for example, the solution may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a suitable sterile aqueous medium may be employed. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the host. The person responsible for administration will, in any event, determine the appropriate dose for the individual host.

Sterile injectable solutions am prepared by incorporating the compositions (e.g., recombinant nucleic acids, rAAVs, pharmaceutical compositions, etc.) in the required amount in the appropriate solvent with various of the other ingredients enumerated herein, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

The compositions (e.g., recombinant nucleic acids, rAAVs, pharmaceutical compositions, etc.) disclosed herein may also be formulated in a neutral or salt form. Pharmaceutically-acceptable salts, include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as injectable solutions, drug-release capsules, and the like.

As used herein “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Supplementary active ingredients can also be incorporated into the compositions. The phrase “pharmaceutically-acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a host.

Delivery vehicles such as liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, may be used for the introduction of the compositions of the present disclosure into suitable host cells. In particular, the rAAV vector delivered transgenes may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like.

Such formulations may be preferred for the introduction of pharmaceutically acceptable formulations of the nucleic acids or the rAAV constructs disclosed herein. The formation and use of liposomes is generally known to those of skill in the art. Recently, liposomes were developed with improved serum stability and circulation half-times (U.S. Pat. No. 5,741,516). Further, various methods of liposome and liposome like preparations as potential drug carriers have been described (U.S. Pat. Nos. 5,567,434; 5,552,157; 5,565,213; 5,738,868 and 5,795,587).

Liposomes have been used successfully with a number of cell types that are normally resistant to transfection by other procedures. In addition, liposomes are free of the DNA length constraints that are typical of viral-based delivery systems. Liposomes have been used effectively to introduce genes, drugs, radiotherapeutic agents, viruses, transcription factors and allosteric effectors into a variety of cultured cell lines and animals. In addition, several successful clinical trials examining the effectiveness of liposome-mediated drug delivery have been completed.

Liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilarmellar vesicles (MLVs). MLVs generally have diameters of from 25 nm to 4 μm. Sonication of MLVs results in the formation of small unilamellar vesicles (SUVs) with diameters in the range of 200 to 500.ANG., containing an aqueous solution in the core.

Alternatively, nanocapsule formulations of the rAAV may be used. Nanocapsules can generally entrap substances in a stable and reproducible way. To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 μm) should be designed using polymers able to be degraded in vivo. Biodegradable polyalkyl-cyanoacrylate nanoparticles that meet these requirements are contemplated for use.

Methods for Treating Rett Syndrome

In some aspects, the disclosure relates to compositions and methods for treating Rett Syndrome. Rett syndrome is a genetic neurological disorder caused by one or more loss of function mutations in the MeCP2 gene, for example as described in Suter et al. J Autism Dev Disord. 2014 March, 44(3): 703-711. In some embodiments, a subject having Rett syndrome has 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more loss of function mutations in MeCP2 gene.

As used herein, the terms “treatment”, “treating”, and “therapy” refer to therapeutic treatment and prophylactic or preventative manipulations. The terms further include ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying causes of symptoms, preventing or reversing causes of symptoms, for example, symptoms associated with a disease caused by a loss of function mutation, for example Rett syndrome. Thus, the terms denote that a beneficial result has been conferred on a subject with a disorder (e.g., Rett syndrome), or with the potential to develop such a disorder. Furthermore, the term “treatment” is defined as the application or administration of an agent (e.g., therapeutic agent or a therapeutic composition) to a subject, or an isolated tissue or cell line from a subject, who may have a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.

Therapeutic agents or therapeutic compositions may include a compound in a pharmaceutically acceptable form that prevents and/or reduces the symptoms of a particular disease (e.g., Rett syndrome). For example a therapeutic composition may be a pharmaceutical composition that prevents and/or reduces the symptoms of Rett syndrome. It is contemplated that the therapeutic composition of the present invention will be provided in any suitable form. The form of the therapeutic composition will depend on a number of factors, including the mode of administration as described herein. The therapeutic composition may contain diluents, adjuvants and excipients, among other ingredients as described herein.

EXAMPLE Example 1: Gene Expression Analysis of Human MeCP2 Isoform e1 In Vitro

Approximately 80% of Rett cases are caused by mutations in the X-linked gene encoding methyl CpG binding protein 2 (MeCP2), a widely expressed epigenetic regulator that is expressed at high levels in mature neurons. Most Rett patients carry a normal and mutant allele of MeCP2. Disease results from random X-chromosome inactivation where ˜50% of neurons are MeCP2 deficient due to inactivation of the normal allele, whereas in the other ˜50% of neurons the mutant allele is silenced and normal expression of wild type MeCP2is retained. The heterogeneity of MeCP2 deficiency in the CNS has important implication, for development of gene therapy approaches for Rett syndrome. In Rett mouse models, the reversibility of neurological phenotypes has been observed after restoration of normal MeCP2 expression in adults. In these transgenic experiments, restored MeCP2 expression was driven from its native genomic locus and activation was achieved in the majority of cells in the brain. However, somatic gene transfer has yet to replicate any of these successes.

Generally, MeCP2 has a very narrow window of safe expression levels, as patients with a duplication of the MeCP2 locus typically present delayed motor and cognitive development as well as severe intellectual impairment. Experiments in transgenic mouse models corroborate this notion, as ectopic expression of MeCP2 is toxic in wild-type animals, but safe and partially effective in ameliorating disease phenotypes of MeCP2-deficient mice when transgene expression starts during embryonic development. Notably, the MeCP2 gene is alternatively spliced to generate two proteins with different N termini, designated as MeCP2-e1 and MeCP2-e2. Patients with MeCP2 locus duplication overexpress both MECP2 isoforms. Therefore, the symptoms in patients with MeCP2 locus duplication and results in transgenic mice may be explained by overexpression of the MeCP2-e2 isoform and timing of transgene expression during development.

Previous AAV9-MeCP2-e1 therapeutic experiments have been focused on neonatal intravascular (IV) or intracerebroventricular (ICV) delivery and in some instances have encountered lethal liver toxicity and hind limb clasping. Furthermore, the age of mice treated in such experiments does not necessarily correspond to that likely to be implemented in most Rett patients, which presumably would be treated after symptom onset (6-18 months). In humans, the primary phase of synaptogenesis occurs in the first 2 years and coincides with a rapid increase in non-CG DNA methylation in neurons, as well as the onset of symptoms in Rett patients. In mice, synaptogenesis occurs between 2 and 4 weeks of age. Therefore, it is critical to examine efficacy and potential toxicity of AAV-MeCP2 gene delivery at relevant developmental stages beyond post-natal day 0-1. Additionally, an important limitation to implementing systemic AAV gene delivery to treat CNS disorders is the transduction of organs other than the brain, such as liver, which is the organ with the highest AAV tropism in the body.

A series of new AAV-MeCP2 vectors that eliminate gene expression in peripheral organs and also self-regulate expression of MeCP2 were designed. Generally, MeCP2 mRNA carries either a short (1.8kb) or long (˜10kb) 3′UTR, with the latter being the preferential isoform expression in brain. The MeCP2 mRNA constructs described in this example comprise an MeCP2 isoform-e1 protein coding sequence and several miRNA regulatory elements (MREs). It was observed that translation of MeCP2 in the CNS is regulated by miR-132 through a homeostatic mechanism involving changes in brain derived neurotrophic factor (BNDF) levels in response to MeCP2 expression (FIG. 1A). Based on this mechanism a series of AAV-MeCP2 vectors with increasing numbers of the miR-132 MREs (e.g., miR-132 binding sites) coupled to a fixed number of MREs for miR-1 and miR-122 (e.g., 3×-miR-1 and 3×-mir-122 binding sites) to de-target AAV gene expression from skeletal muscle and liver (FIG. 1B).

A series of in vitro experiments were carried out. Briefly, HEK293T cells were transfected with 30,000 gc/cell of AAV2-MeCP2 for four days. FIGS. 2A-2B show effective expression of AAV2-MeCP2 in HEK293T cells, as measured by Western blot (IG. 2A) and normalized protein expression assay (FIG. 2B). FIG. 2C shows a toxicity profile of 293T cells transduced with AAV2-MeCP2 for four days at a dose of 30,000 gc/cell.

A dose response study in mouse primary cortical neurons showed comparable effects on cell survival for AAV-GFP and AAV-MeCP2 vectors (FIG. 3A), indicating that expression of myc-tagged human MeCP2 from a short mouse MeCP2 promoter (−223 to +56) is non-toxic to primary neurons in culture. In addition, it was observed that MeCP2-myc protein levels were inversely proportional to the number of miR-132 MREs (e.g., miR-132 binding sites) present in the MeCP2-myc transcript (FIG. 3B). FIG. 3C shows miR-132 expression in response to AAV2-MeCP2 five days after AAV infection.

Example 2: Gene Expression Analysis of Human MeCP2 Isoform e1 in Wild-Type Mice Following Systemic Delivery of AAV-MeCP2

To extend the in vitro observations demonstrating the ability to titer MeCP2 levels by insertion of miR-132 target sequences described in Example 1, post-natal day 1 wild-type mice were injected via the facial vein (e.g., intracranial injection) with AAV encoding the e1 isoform of human MeCP2 containing 0, 1×, 2×, or 3× miR-132 target sequences. Gene expression analysis of brain tissue indicated that MeCP2 levels are inversely proportional to the number of miR132 target sequences (FIGS. 4A-4C).

In some embodiments, systemic administration of some AAV serotypes can transduce tissues outside of the central nervous system, and elevated expression of MeCP2 in liver, cardiac and skeletal tissues has been observed to be associated with detrimental physiological consequences. To minimize heart and liver transduction of MeCP2, AAV-MeCP2 vectors described by the disclosure contain at least one miR-1 (e.g., 3×-miR-1) and at least one miR-122 (e.g., 3×-miR-122) target sequence (e.g., binding sites) to de-target MeCP2 expression from the heart and liver, respectively, qRT-PCR analysis using primers against e2 human MeCP2 (which was undetectable), and e1 and e2 mouse MeCP2 (which did not change) were performed. Gene expression analysis of heart and liver tissue from wild-type animals indicated MeCP2 is effectively de-targeted from the heart and liver, as evidenced by substantially reduced expression compared to the brain (FIG. 4A and FIG. 5).

Example 3: Therapeutic Efficacy and Safety of Self-Regulating AAV-MeCP2 Vectors

Therapeutic efficacy and safety of AAV-MeCP2-e| vectors is examined in mice. In some embodiments, AAV-PHP.B capsid protein is used, as this capsid has improved neuronal transduction efficiency. Mecp2-null mice (Mecp2tm1.1Bind/J; Male−/y and female+/−) at 4 weeks of age are treated by systemic administration of AAV-PHP.B-MeCP2-e1 vectors carrying different MRE cassettes (e.g., at vector doses of 1E11, 3E11, 1E12 vg/mouse) and body weight and phenotypic scores are monitored every two weeks. As controls. MeCP2/Mecp2tm1.1Bind mice injected with vehicle and wild type mice are used. A subset of mice in each cohort (n=8; 4 males and 4 females) are sacrificed at 8 weeks post-injection and MeCP2 expression quantified by western blot and compared across groups. Transduction efficiency in the brain is assessed by double immunofluorescence staining for MeCP2 and neurons (using the neuronal marker NeuN) and quantification of transduced neurons (MeCP2+, NeuN+) in cortex, striatum, thalamus, hippocampus and cerebellum is performed. The levels of PSD-95 are assessed by western blot and immunostaining of brain sections: PSD-95 is a key scaffold protein in synaptic maturation whose levels are decreased in brains from MeCP2-null mice.

To perform vector biodistribution analysis, genomic DNA is isolated from different regions of the CNS and peripheral organs and analyzed by digital PCR. Another subset of animals in each cohort (n=16; 8 males and 8 females) is used for survival and longitudinal analysis of behavioral (e.g., open field; social interaction) and motor performance (e.g., rotarod, grid walk) as well as whole body plethismography to assess breathing patterns and apnea characteristic of MeCP2-null mice. Endpoint studies are the same as at 8 weeks after treatment. Safety of the vectors is also assessed in wild type mice in a dose escalation study using doses identical to those indicated above with endpoints at 7, 30, 90 and 180 days to assess the CNS and peripheral tissues for evidence of toxicity. AAV vector biodistribution and MeCP2 expression are assessed as well.

Example 4: Characterization of Changes in the Genome/Transcriptome of Transduced Neurons after AAV-MeCP2 Gene Transfer at Different Stages of Nervous System Development

A key aspect in the development of a safe and effective gene therapy approach for Rett is to characterize in detail the impact of de novo expression of MeCP2 on the epigenetic landscape and transcriptomic profile of transduced neurons. For this purpose. AAV-PHP.B-MeCP2-e1 vectors carrying an IRES-GFP cassette are produced, and allow isolation of transduced GFP+ cells from brain, cerebellum and spinal cord by either FACS or laser capture microdissection followed by whole genome bisulfite sequencing (MethylC-Seq), small RNA-seq (microRNAs), and RNA-Seq (mRNAs and non-coding RNAs). MeCP2−/y males. MeCP2−/+ females and wild-type controls (males and females) receive a systemic injection of AAV-PHP.B-MeCP2-e1-IRES-GFP, control vector (without MeCP2 cDNA) and vehicle at day 1, 7, 14, and 28, as well as at 12 weeks of age at an optimal dose. Mice (n=8; 4 males and 4 females) are euthanized at 1 or 3 months after injection to assess the parameters indicated above. Information on microRNAs that are overexpressed in response to MeCP2 expression is used to established additional layers of gene expression regulation in addition to that based on miR-132.

Example 5: Contribution of MeCP2 Isoforms to Therapeutic Success or Onset of Neurological Symptoms as a Function of Intervention at Different Stages of Development

MeCP2 expression of 1.6- to 6-fold above physiologically normal levels has been observed to cause neurological symptoms both in patients with MeCP2 locus duplication (˜2-fold above normal) and transgenic mouse models. The other commonality between patients and transgenic mouse models is that both overexpress the MeCP2-e2 isoform, which unlike the e1 isoform appears to be toxic to primary neurons in culture. In some embodiments, this toxic effect is eliminated by co-expression of FoxG1, which is another gene where mutations are associated with Rett syndrome. In some embodiments, co-expression of FoxG1 with MeCP2 is an additional mechanism to control the side effects associated with MeCP2 overexpression. Therapeutic, safety and epigenomic/transcriptomic experiments with AAV-PHP.B vectors encoding MeCP-e1, MeCP2-e2, MeCP2-e2 and FoxG1, or FoxG1 alone in are conducted MeCP2−/y males, MeCP2+/− females and wild-type age matched controls.

SEQUENCES >SEQ ID NO: 1; human MeCP2 isoform el amino acid sequence (NM_001110792) MAAAAAAAPSGGGGGGBEERLEEKSEDQDLQGLKDKPLKFKKVKKDKKEEKEGKHEP VQPSAHHSAEPAEAGKAETSEGSGSAPAVPEASASPKQRRSIIRDRGPMYDDPTLPEGW TRKIKQRKSGRSAGKYDVYLINPQGKAFRSKVELIAYFEKVGDTSLDPNDFDFTVTGRG SPSRREQKPPKKFKSPKAPGTGRGRGRPKGSGTTRPKAATSEGVQVKRVLEKSPGKLLV KMPFQTSPGGKAEGGGATTSTQVMVIKRPGRKRKAEADPQAIPKKRGRKPGSVVAAA AAEAKKKAVKESSIRSVQETVLPIKKRKTRETVSIEVKEVVKPLLVSTLGEKSGKGLKTC KSPGRKSKESSPKGRSSSASSPPKKEHHHHHHHSESPKAPVPLLPPLPPPPPEPESSEDPTS PPEPQDLSSSVCKEEKMPRGGSLESDGCPKEPAKTQPAVATAATAAEKYKHRGEGERK DIVSSSMPRPNREEPVDSRTPVTERVS >SEQ ID NO: 2; human MeCP2 isoform e2 amino acid sequence (NM_004992) MVAGMLGLREEKSEDQDLQGLKDKPLKFKKVKKDKKEEKEGKHEPVQPSAHHSAEPA EAGKAETSEGSGSAPAVPEASASPKQRRSIIRDRGPMYDDPTLPEGWTRKLKQRKSGRS AGKYDVYLINPQGKAFRSKVELIAYFEKVGDTSLDPNDFDFTVTGRGSPSRREQKPPKK PKSPKAPGTGRGRGRPKGSGTTRPKAATSEGVAVKRVLEKSPGKLLVKMPFQTSPGGK AEGGGATTSTQVMVIKRPGRKRKAEADPQAIPKKRGRKPGSVVAAAAAEAKKKAVKE SSIRSVQETVLPIKKRKTRETVSIEVKEVVKPLLVSTLGEKSGKGLKTCKSPGRKSKESSP KGRSSSASSPPKKEHHHHHHHSESPKAPVPLLPPLPPPPPEPESSEDPTSPPEPQDLSSSVC KEEKMPRGGSLESDGCPKEPAKTQPAVATAATAAEKYKHRGEGERKDIVSSSMPRPNR EEPVDSRIPVTERVS >SEQ ID NO: 3; mouse MeCP2 promoter DNA sequence AATTGAGGGCGTCACCGCTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAA GGGTAATCTCGACAAAGAGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCA CACAGGCTGGTCGGGAGGGGGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCG GTT >SEQ ID NO: 4; miR-122 binding site DNA sequence ACAAACACCATTGTCACACTCCA >SEQ ID NO: 5; miR-1 binding site DNA sequence ATACATACTTCTTTACATTCCA >SEQ ID NO: 6; miR-132 binding site DNA sequence CGACCATGGCTGTAGACTGTTA >SEQ ID NO: 7; MeCP2 in vitro construct nucleic acid sequence (scAAV-Mec229- hMeCP2-miR132(1x) miR122-1(3x) plasmid) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GCTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCGCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC AGCCCACCACTCTGCTGAGCCCCCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGCTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GCGGTTGCCACCGCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG GACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACTCGAGATACATACTTCTTTACAT TCCAATACATACTTCTTTACATTCCAATACATACTTCTTTACATTCCACCATGGACTA GTACAAACACCATTGTCACACTCCAACAAACACCATTGTCACACTCCAACAAACAC CATTGTCACACTCCAGCGGCCGCTTCGATCCGATCTTTTTCCCTCTGCCAAAAATTAT GGGGACATCATGAAGCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATT TTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGCCTAGGTAGATAA GTAGCATGGCGGGTTAATCATTAACTACAAGGAACCCCTAGTGATGGAGTTGGCCA CTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCGAC CCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAGCCTTAATTAA CCTAATTCACTGGCCGTCGTTTTACAACGTCGTGACTGGGAAAACCCTGGCGTTACC CAACTTAATCGCCTTGCAGCACATCCCCCTTTCGCCAGCTGGCGTAATAGCGAAGAG GCCCGCACCGATCCCCCTTCCCAACAGTTGCGCAGCCTGAATGGCGAATGGGACGC GCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGCGTGACCG CTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCGC CACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCG ATTTAGTGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACG TAGTGGGCCATGGCCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTT CTTTAATAGTGGACTCTTGTTCCAAACTGGAACAACACTCAACCCTATCTCGGTCTA TTCTTTTGATTTATAAGGGATTTTGCCGATTTCGGCCTATTGGTTAAAAAATGAGCT GATTTAACAAAAATTTAACGCGAATTTTAACAAAATATTAACGCTTACAATTTAGGT GGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTTTATTTTTCTAAATACAT TCAAATATGTATCCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAATATTGA AAAAGGAAGAGTATGAGTATTCAACATTTCCGTGTCGCCCTTATTCCCTTTTTTGCG GCATTTTGCCTTCCTGTTTTTGCTCACCCAGAAACGCTGGTGAAAGTAAAAGATGCT GAAGATCAGTTGGGTGCACGAGTGGGTTACATCGAACTGGATCTCAACAGCGGTAA GATCCTTGAGAGTTTTCGCCCCGAAGAACGTTTTCCAATGATGAGCACTTTTAAAGT TCTGCTATGTGGCGCGGTATTATCCCGTATTGACGCCGGGCAAGAGCAACTCGGTCG CCGCATACACTATTCTCAGAATGACTTGGTTGAGTACTCACCAGTCACAGAAAAGC ATCTTACGGATGGCATGACAGTAAGAGAATTATGCAGTGCTGCCATAACCATGAGT GATAACACTGCGGCCAACTTACTTCTGACAACGATCGGAGGACCGAAGGAGCTAAC CGCTTTTTTGCACAACATGGGGGATCATGTAACTCGCCTTGATCGTTGGGAACCGGA GCTGAATGAAGCCATACCAAACGACGAGCGTGACACCACGATGCCTGTAGCAATGG CAACAACGTTGCGCAAACTATTAACTGGCGAACTACTTACTCTAGCTTCCCGGCAAC AATTAATAGACTGGATGGAGGCGGATAAAGTTGCAGGACCACTTCTGCGCTCGGCC CTTCCGGCTGGCTGGTTTATTGCTGATAAATCTGGAGCCGGTGAGCGTGGGTCTCGC GGTATCATTGCAGCACTGGGGCCAGATGGTAAGCCCTCCCGTATCGTAGTTATCTAC ACGACGGGGAGTCAGGCAACTATGGATGAACGAAATAGACAGATCGCTGAGATAG GTGCCTCACTGATTAAGCATTGGTAACTGTCAGACCAAGTTTACTCATATATACTTT AGATTGATTTAAAACTTCATTTTTAATTTAAAAGGATCTAGGTGAAGATCCTTTTTG ATAATCTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACC CCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCT GCTTGCAAACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAG CTACCAACTCTTTTTCCGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACT GTTCTTCTAGTGTAGCCGTAGTTAGGCCACCACTTCAAGAACTCTGTAGCACCGCCT ACATACCTCGCTCTGCTAATCCTGTTACCAGTGGCTGCTGCCAGTGGCGATAAGTCG TGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGGATAAGGCGCAGCGGTCGGG CTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGACCTACACCGAAC TGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGAAA GGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAG CTTCCAGGGGGAAACGCCTGGTATGTTTATAGTCCTGTCGGGTTTCGCCACCTCTGA CTTGAGCGTCGATTTTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGC CAGCAACGCGGCCTTTTTACGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTC TTTCCTGCGTTATCCCCTGATTCTGTGGATAACCGTATTACCGCCTTTGAGTGAGCTG ATACCGCTCGCCGCAGCCGAACGACCGAGCGCAGCGAGTCAGTGAGCGAGGAAGC GGAAGAGCGCCCAATACGCAAACCGCCTCTCCCCGCGCGTTGGCCGATTCATTAAT GCAGCTGGCACGACAGGTTTCCCGACTGGAAAGCGGGCAGTGAGCGCAACGCAATT AATGTGAGTTAGCTCACTCATTAGGCACCCCAGGCTTTACACTTTATGCTTCCGGCT CGTATGTTGTGTGGAATTGTGAGCGGATAACAATTTCACACAGGAAACAGCTATGA CCATGATTACGCCAGATTTAATTAAGGCCTTAATTAGG >SEQ ID NO: 8; MeCP2 in vitro construct nucleic acid sequence (scAAV-Mex229- hMeCP2-miR132(2x) miR122-1(3x) plasmid) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GCTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCGCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC AGCCCACCACTCTGCTGAGCCCCCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGCTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GCGGTTGCCACCGCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG GACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACGACCATGGCTGTAGACTGTTACT CGAGATACATACTTCTTTACATTCCAATACATACTTGTTTACATTCCAATACATACTT CTTTACATTCCACCATGGACTAGTACAAACACCATTGTCACACTCCAACAAACACCA TTGTCACACTCCAACAAACACCATTGTCACACTCCAGCGGCCGCTTCGATCCGATCT TTTTCCCTCTGCCAAAAATTATGGGGACATCATGAAGCCCCTTGAGCATCTGACTTC TGGCTAATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTC TCACTCGGCCTAGGTAGATAAGTAGCATGGCGGGTTAATCATTAACTACAAGGAAC CCCTAGTGATGGAGTTGGCCACTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCG GGCGACCAAAGGTCGCCCGACCCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAG CGAGCGCGCAGCCTTAATTAACCTAATTCACTGGCCGTCGTTTTACAACGTCGTGAC TGGGAAAACCCTGGCGTTACCCAACTTAATCGCCTTGCAGCACATCCCCCTTTCGCC ACCTGGCGTAATAGCGAAGAGGCCCGCACCGATCGCCCTTCCCAACAGTTGCGCAG CCTGAATGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGG TGGTTACGCGCAGCGTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCG CTTTCTTCCCTTCCTTTCTCGCCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCG GGGGCTCCCTTTTAGGGTTCCGATTTAGTGCTTTACGGCACCTCGACCCCAAAAAACT TGATTAGGGTGATGGTTCACGTAGTGGGCCATCGCCCTGATAGACGGTTTTTCGCCC TTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGTTCCAAACTGGAACAAC ACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCCGATTTCGGCC TATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAACAAAAT ATTAACGCTTACAATTTAGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATT TGTTTATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAGACAATAACCCTGA TAAATGCTTCAATAATATTGAAAAAGGAAGAGTATGAGTATTCAACATTTCCGTGTC GCCCTTATTCCCTTTTTTGCGGCATTTTGCCTTCCTGTTTTTGCTCACCCAGAAACGC TGGTGAAAGTAAAAGATGCTGAAGATCAGTTGGGTGCACGAGTGGGTTACATCGAA CTGGATCTCAACAGCGGTAAGATCGTTGAGAGTTTTCGCCCCGAAGAACGTTTTCCA ATGATGAGCACTTTTAAAGTTCTGCTATGTGGCGCGGTATTATCCCGTATTGACGCC GGGCAAGAGCAACTCGGTCGCCGCATACACTATTCTCAGAATGACTTGGTTGAGTA CTCACCAGTCACAGAAAAGCATGTTACGGATGGCATGACAGTAAGAGAATTATGCA GTGCTGCCATAACCATGAGTGATAACACTGCGGCCAACTTACTTCTGACAACGATC GGAGGACCGAAGGAGCTAACCGCTTTTTTGCACAACATGGGGGATCATGTAACTCG CCTTGATCGTTGGGAACCGGAGCTGAATGAAGCCATACCAAACGACGAGCGTGACA CCACGATGCCTGTAGCAATGGCAACAACGTTGCGCAAACTATTAACTGGCGAACTA CTTACTCTAGCTTCCCGGCAACAATTAATAGACTGGATGGAGGCGGATAAAGTTGC AGGACCACTTCTGCGCTCGGCCCTTCCGGCTGGCTGGTTTATTGCTGATAAATCTGG AGCCGGTGAGCGTGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGTAAGC CCTCCCGTATCGTAGTTATCTACACGACGGGGAGTCAGGCAACTATGGATGAACGA AATAGACAGATCGCTGAGATAGGTGCCTCACTGATTAAGCATTGGTAACTGTCAGA CCAAGTTTACTCATATATACTTTAGATTGATTTAAAACTTCATTTTTAATTTAAAAGG ATCTAGGTGAAGATCCTTTTTGATAATCTCATGACCAAAATCCCTTAACGTGAGTTT TCGTTCCACTGAGCGTCAGACCCCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCT TTTTTTCTGCGCGTAATCTGCTGCTTGCAAACAAAAAAACCACCGCTACCAGCGGTG GTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTCCGAAGGTAACTGGCTTCAGC AGAGCGCAGATACCAAATACTGTTCTTCTAGTGTAGCCGTAGTTAGGCCACCACTTC AAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCAGTGGCT GCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACC GGATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACACCCCAGCTTGG AGCGAACGACCTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCC ACGGTTCCCGAAGGGAGAAAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAA CAGGAGAGGGCACGAGGGAGCTTCCAGGGGGAAACGCCTGGTATCTTTATAGTCCT GTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATTTTTGTGATGCTCGTCAGGGGGG CGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGGTTCCTGGCCTTTTGC TGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGGATAACCGTA TTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAGC GAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCCAATACGCAAACCGCCTCTCCCCG CGCGTTGGCCGATTCATTAATGCAGCTGGCACGACAGGTTTCCCGACTGGAAAGCG GGCAGTGAGCGCAACGCAATTAATGTGAGTTAGCTCACTCATTAGGCACCCCAGGC TTTACACTTTATGCTTCCGGCTCGTATGTTGTGTGGAATTGTGAGCGGATAACAATT TCACACAGGAAACAGCTATGACCATGATTACGCCAGATTTAATTAAGGCCTTAATT AGG >SEQ ID NO: 9; MeCP2 in vitro construct nucleic acid sequence (scAAV-Mec229- hMeCP2-miR132(3x) miR122-1(3x) plasmid) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GGTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCGCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC AGCCCACCACTCTGCTGAGCCCGCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGCTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GCGGTTGCCACCGCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG GACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACGACCATGGCTGTAGACTGTTAC CACCATGGCTGTAGACTGTTACTCGAGATACATACTTCTTTACATTCCAATACATAC TTCTTTACATTCCAATACATACTTCTTTACATTCCACCATGGACTAGTACAAACACC ATTGTCACACTCCAACAAACACCATTGTCACACTCCAACAAACACCATTGTCACACT CCAGCGGCCGCTTCGATCCGATCTTTTTCCCTCTGCCAAAAATTATGGGGACATCAT GAAGCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATTTTCATTGCAAT AGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGCCTAGGTAGATAAGTAGCATGGCG CGTTAATCATTAACTACAAGGAACCCCTAGTGATGGAGTTGGCCACTCCCTCTCTGC GCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCGACGCCCGGGCTTT GCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAGCCTTAATTAACCTAATTCACT GGCCGTCGTTTTACAACCTCGTGACTGGGAAAACCCTGGCGTTACCCAACTTAATCG CCTTGCAGCACATCCCCCTTTCGCCAGCTGGCGTAATAGCGAAGAGGCCCGCACCG ATCGCCCTTCCCAACAGTTGCGCAGCCTGAATGGCGAATGGGACGCGCCCTGTAGC GGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGCGTGACCGCTACACTTGC CAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCGCCACGTTCGCC GGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAGTGCT TTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCC ATCGCCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGT GGACTCTTGTTCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATT TATAAGGGATTTTGCCGATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAA AATTTAACGCGAATTTTAACAAAATATTAACGCTTACAATTTAGGTGGCACTTTTCG GGGAAATGTGCGCGGAACCCCTATTTGTTTATTTTTCTAAATACATTCAAATATGTA TCCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAATATTGAAAAAGGAAGA GTATGAGTATTCAACATTTCCGTGTCGCCCTTATTCCCTTTTTTGCGGCATTTTGCCT TCCTGTTTTTGCTCACCCAGAAACGCTGGTGAAAGTAAAAGATGCTGAAGATCAGTT GGGTGCACGAGTGGGTTACATCGAACTGGATCTCAACAGCGGTAAGATCCTTGAGA GTTTTCGCCCCGAAGAACGTTTTCCAATGATGAGCACTTTTAAAGTTCTGCTATGTG GGGCGGTATTATCCCGTATTGACGCCGGGCAAGAGCAACTCGGTCGCCGCATACAC TATTCTCAGAATGACTTGGTTGAGTACTCACCAGTCACAGAAAAGCATCTTACGGAT GGCATGACAGTAAGAGAATTATGCAGTGCTGCCATAACCATGAGTGATAACACTGC GGCCAACTTACTTCTGACAACGATCGGAGGACCGAAGGAGCTAACCGCTTTTTTGC ACAACATGGGGGATCATGTAACTCGCCTTGATCGTTGGGAACCGGAGCTGAATGAA GCCATACCAAACGACGAGCGTGACACCACGATGCCTGTAGCAATGGCAACAACGTT GCGCAAACTATTAACTGGCGAACTACTTACTCTAGCTTCCCGGCAACAATTAATAGA CTGGATGGAGGCGGATAAAGTTGCAGGACCACTTCTGCGCTCGGCCCTTCCGGCTG GCTGGTTTATTGCTGATAAATCTGGAGCCGGTGAGCGTGGGTCTCGCGGTATCATTG CAGCACTGGGGCCAGATGGTAAGCCCTCCCGTATCGTAGTTATCTACACGACGGGG AGTCAGGCAACTATGGATGAACGAAATAGACAGATCGCTGAGATAGGTGCCTCACT GATTAAGCATTGGTAACTGTCAGACCAAGTTTACTCATATATACTTTAGATTGATTT AAAACTTCATTTTTAATTTAAAAGGATCTAGGTGAAGATCCTTTTTGATAATCTCAT GACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCCCGTAGAAA AGATCAAAGGATGTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAAA CAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACT CTTTTTCCGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTTCTTCTA GTGTAGCCGTAGTTAGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTC GCTCTGCTAATCCTGTTACCAGTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACC GGGTTGGACTCAAGACGATAGTTACCGGATAAGGCGCAGCGGTCGGGCTGAACGG GGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGACCTACACCGAACTGAGATAC CTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGAAAGGCGGACA GGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAGG GGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCG TCGATTTTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACG CGGCCTTTTTACGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGC GTTATCCCCTGATTCTGTGGATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGC TCGCCGCAGCCGAACGACCGAGCGCAGCGAGTCAGTGAGCGAGGAAGCGGAAGAG CGCCCAATACGCAAACCGCCTCTCCCCGCGCGTTGGCCGATTCATTAATGCAGCTGG CACGACAGGTTTCCCGACTGGAAAGCGGGCAGTGAGCGCAACGCAATTAATGTGAG TTAGCTCACTCATTAGGCACCCCAGGCTTTACACTTTATGCTTCCGGCTCGTATGTTG TGTGGAATTGTGAGCGGATAACAATTTCACACAGGAAACAGCTATGACCATGATTA CGCCAGATTTAATTAAGGCCTTAATTAGG >SEQ ID NO: 10; MeCP2 in vivo construct nucleic acid sequence (scAAV-Mec229- bMeCP2-miR132(1x) miR 122-1(3x) vector genome) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GCTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCCCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC ACCCCACCACTCTGCTGAGCCCGCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGGTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GCGGTTGCCACCGCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG CACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACTCGAGATACATACTTCTTTACAT TCCAATACATACTTCTTTACATTCCAATACATACTTCTTTACATTCCACCATGGACTA GTACAAACACCATTGTCACACTCCAACAAACACCATTGTCACACTCCAACAAACAC CATTGTCACACTCCAGCGGCCGCTTCGATCCGATCTTTTTCCCTCTGCCAAAAATTAT GGGGACATCATGAAGCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATT TTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGCCTAGGTAGATAA GTAGCATGGCGGGTTAATCATTAACTACAAGGAACCCCTAGTGATGGAGTTGGCCA CTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCGAC GCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAG >SEQ ID NO: 11; MeCP2 in vivo construct nucleic acid sequence (scAAV-Mec229- hMeCP2-miR132(2x) miR122-1(3x) vector genome) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GCTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCCCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC AGCCCACCACTCTGCTGAGCCCGCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGGTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GCGGTTGCCACCCCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG GACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACGACCATGGCTGTAGACTGTTACT CGAGATACATACTTGTTTACATTCCAATACATACTTCTTTACATTCCAATACATACTT CTTTACATTCCACCATGGACTAGTACAAACACCATTGTCACACTCCAACAAACACCA TTGTCACACTCCAACAAACACCATTGTCACACTCCAGCGGCCGCTTCGATCCGATCT TTTTCCCTCTGCCAAAAATTATGGGGACATCATGAAGCCCCTTGAGCATCTGACTTC TGGCTAATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTC TCACTCGGCCTAGGTAGATAAGTAGCATGGCGGGTTAATCATTAACTACAAGGAAC CCCTAGTGATGGAGTTGGCCACTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCG GGCGACCAAAGGTCGCCCGACGCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAG CGAGCGCGCAG >SEQ ID NO: 12; MeCP2 in vivo construct nucleic acid sequence (scAAV-Mec229- hMeCP2-miR132(3x) miR 122-1(3x) vector genome) CTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCAAAGCCCGGGCGTCGGGCGAC CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGTAGCCAT GCTCTAGGAAGATCAATTCGGTACAATTCACGCGTCGACAATTGAGGGCGTCACCG CTAAGGCTCCGCCCCAGCCTGGGCTCCACAACCAATGAAGGGTAATCTCGACAAAG AGCAAGGGGTGGGGCGCGGGCGCGCAGGTGCAGCAGCACACAGGCTGGTCGGGAG GGCGGGGCGCGACGTCTGCCGTGCGGGGTCCCGGCATCGGTTGCGCGCGCGCTCCC TCCTCTCGGAGAGAGGGCTGTGGTAAAACCCGTCCGGAAAATGGCTGCAGCCGCTG CCGCAGCGCCGAGCGGCGGAGGTGGCGGTGGCGAGGAGGAGAGACTGGAAGAAAA GTCAGAAGACCAGGACCTCCAGGGCCTCAAGGACAAACCCCTCAAGTTTAAAAAGG TGAAGAAAGATAAGAAAGAAGAGAAAGAGGGCAAGCATGAGCCCGTGCAGCCATC AGCCCACCACTCTGCTGAGCCCGCAGAGGCAGGCAAAGCAGAGACATCAGAAGGG TCAGGCTCCGCCCCGGCTGTGCCGGAAGCTTCTGCCTCCCCCAAACAGCGGCGCTCC ATCATCCGTGACCGGGGACCCATGTATGATGACCCCACCCTGCCTGAAGGCTGGAC ACGGAAGCTTAAGCAAAGGAAATCTGGACGCTCTGCTGGGAAGTATGATGTGTATT TGATCAATCCCCAGGGAAAAGCCTTTCGCTCTAAAGTGGAGTTGATTGCGTACTTCG AAAAGGTAGGCGACACATCCCTGGACCCTAATGATTTTGACTTCACGGTAACTGGG AGAGGGAGCCCCTCCCGGCGAGAGCAGAAACCACCTAAGAAGCCCAAATCTCCCA AAGCTCCAGGAACTGGCAGAGGTCGGGGACGCCCCAAAGGGAGCGGCACCACGAG ACCCAAGGCAGCTACGTCAGAGGGTGTGCAGGTGAAAAGGGTCCTGGAGAAAAGT CCTGGGAAGCTCCTTGTCAAGATGCCTTTTCAAACTTCGCCAGGGGGCAAGGCTGA GGGGGGTGGGGCCACCACATCCACCCAGGTCATGGTGATCAAACGCCCCGGCAGGA AGCGAAAAGCTGAGGCAGACCCTCAGGCCATTCCCAAGAAACGGGGTCGAAAGCC GGGGAGTGTGGTGGCAGCCGCTGCCGCCGAGGCCAAAAAGAAAGCCGTGAAGGAG TCTTCTATCCGATCTGTGCAGGAGACCGTACTCCCCATCAAGAAGCGCAAGACCCG GGAGACGGTCAGCATCGAGGTCAAGGAAGTGGTGAAGCCCCTGCTGGTGTCCACCC TCGGTGAGAAGAGCGGGAAAGGACTGAAGACCTGTAAGAGCCCTGGGCGGAAAAG CAAGGAGAGCAGCCCCAAGGGGCGCAGCAGCAGCGCCTCCTCACCCCCCAAGAAG GAGCACCACCACCATCACCACCACTCAGAGTCCCCAAAGGCCCCCGTGCCACTGCT CCCACCCCTGCCCCCACCTCCACCTGAGCCCGAGAGCTCCGAGGACCCCACCAGCC CCCCTGAGCCCCAGGACTTGAGCAGCAGCGTCTGCAAAGAGGAGAAGATGCCCAG AGGAGGCTCACTGGAGAGCGACGGCTGCCCCAAGGAGCCAGCTAAGACTCAGCCC GGGGTTGCCACCGCCGCCACGGCCGCAGAAAAGTACAAACACCGAGGGGAGGGAG AGCGCAAAGACATTGTTTCATCCTCCATGCCAAGGCCAAACAGAGAGGAGCCTGTG GACAGCCGGACGCCCGTGACCGAGAGAGTTAGCGAGCAGAAGCTGATCTCAGAGG AGGACCTGTGACGACCATGGCTGTAGACTGTTACGACCATGGCTGTAGACTGTTAC GACCATGGCTGTAGACTGTTACTCGAGATACATACTTCTTTACATTCCAATACATAC TTCTTTACATTCCAATACATACTTCTTTACATTCCACCATGGACTAGTACAAACACC ATTGTCACACTCCAACAAACACCATTGTCACACTCCAACAAACACCATTGTCACACT CCAGCGGCCGCTTCGATCCGATCTTTTTCCCTCTGCCAAAAATTATGGGGACATCAT GAAGCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATTTTCATTGCAAT AGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGCCTAGGTAGATAAGTAGCATGGCG GGTTAATCATTAACTACAAGGAACCCCTAGTGATGGAGTTGGCCACTCCCTCTCTGC GCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCGACGCCCGGGCTTT CCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAG >SEQ ID NO: 13; AAV2 capsid amino acid sequence MAADGYLPDWLEDTLSEGIRQWWKLKPGPPPPKPAERHKDDSRGLVLPGYKYLGPFN GLDKGEPVNEADAAALEHDKAYDRQLDSGDNPYLKYNHADAEFQERLKEDTSFGGNL GRAVFQAKKRVLEPLGLVEEPVKTAPGKKRPVEHSPVEPDSSSGTGKAGQQPARKRLN FGQTGDADSVPDPQPLGQPPAAPSGLGTNTMATGSGAPMADNNEGADGVGNSSGNWH CDSTWMGDRVITTSTRTWALPTYNNHLYKQISSQSGASNDNHYFGYSTPWGYFDFNRF HCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTQNDGTTTIANNLTSTVQVFTDS EYQLPYVLGSAHQGCLPPFPADVFMVPQYGYLTLNNGSQAVGRSSFYCLEYFPSQMLR TGNNFTFSYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLSRTNTPSGTTTQSRLQFSQ AGASDIRDQSRNWLPGPCYRQQRVSKTSADNNNSEYSWTGATKYHLNGRDSLVNPGP AMASHKDDEEKFFPQSGVLIFGKQGSEKTNVDIEKVMITDEEEIRTTNPVATEQYGSVST NLQRGNRQAATADVNTQGVLPGMVWQDRDVYLQGPIWAKIPHTDGHFHPSPLMGGF GLKHPPPQILIKNTPVPANPSTTFSAAKFASFITQYSTGQVSVEIEWELQKENSKRWNPEI QYTSNYNKSVNVDFTVDINGVYSEPRPIGTRYLTRNL >SEQ ID NO: 14; AAV9 capsid amino acid sequence MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLGPG NGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDTSFGG NLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQPAKKR LNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMASGGGAPVADNNEGADGVGSSSGNW HCDSQWLGDRVITTSTRTWALPTYNNHLYKQISNSTSGGSSNDNAYFGYSTPWGYFDF NRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTDNNGVKTIANNLTSTVQV FTDSDYQLPYVLGSAHEGCLPPFPADVFMIPQYGYLTLNDGSQAVGRSSFYCLEYFPSQ MLRTGNNFQFSYEFENVPFHSSYAHSQSLDRLMNPLIDQYLYYLSKTINGSGQNQQTLK FSVAGPSNMAVQGRNYIPGPSYRQQRVSTTVTQNNNSEFAWPGASSWALNGRNSLMN PGPAMASHKEGEDRFFPLSGSLIFGKQGTGRDNVDADKVMITNEEEIKTTNPVATESYG QVATNHQSAQAQAQTGWVQNQGILPGMVWQDRDVYLQGPIWAKIPHTDGNFHPSPL MGGFGMKHPPPQILIKNTPVPADPPTAFNKDKLNSFITQYSTGQVSVEIEWELQKENSKR WNPEIQYTSNYYKSNNVEFAVNTEGVYSEPRPIGTRYLTRNL >SEQ ID NO: 15; AAV-PHP.B capsid amino acid sequence MAADGYLPDWLEDNLSEGIREWWALKPGAPQPKANQQHQDNARGLVLPGYKYLGPG NGLDKGEPVNAADAAALEHDKAYDQQLKAGDNPYLKYNHADAEFQERLKEDTSFGG NLGRAVFQAKKRLLEPLGLVEEAAKTAPGKKRPVEQSPQEPDSSAGIGKSGAQPAKKR LNFGQTGDTESVPDPQPIGEPPAAPSGVGSLTMASGGGAPVADNNEGADGVGSSSGNW HCDSQWLGDRVITTSTRTWALPTYNNHLYKQISNSTSGGSSNDNAYFGYSTPWGYFDP NRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTDNNGVKTIANNLTSTVQV FTDSDYQLPYVLGSAHEGCLPPFPADVFMIPQYGYLTLNDGSQAVGRSSFYCLEYFPSQ MLRTGNNFQFSYEFENVPFHSSYAHSQSLDRLMNPLIDQYLYYLSRTINGSGQNQQTLK FSVAGPSNMAVQGRNYIPGPSYRQQRVSTTVTQNNNSEFAWPGASSWALNGRNSLMN PGPAMASHKEGEDRFFPLSGSLIFGKQGTGRDNVDADKVMITNEEEIKTTNPVATESYG QVATNHQSAQTLAVPFKAQAQTGWVQNQGILPGMVWQDRDVYLQGPIWAKIPHTDG NFHPSPLMGGFGMKHPPPQILIKNTPVPADPPTAFNKDKLNSFITQYSTGQVSVEIEWEL QKENSKRWNPEIQYTSNYYKSNNVEFAVNTEGVYSEPRPIGTRYLTRNL

Claims

1. A method of engineering a transgene, the method comprising:

(a) selecting a first gene encoding a first product in a cell;
(b) selecting an miRNA, the expression of which is positively regulated by the first product in the cell; and,
(c) engineering a transgene that expresses a transcript having a coding region encoding the first product and a 3′-non-coding region comprising one or more binding sites for the miRNA.

2. A method of engineering a transgene, the method comprising:

(a) selecting a first gene encoding a first product in a cell;
(b) selecting a second gene encoding a second product in the cell;
(c) determining that expression of the second product is positively regulated by the first product in the cell;
(d) selecting an miRNA;
(e) determining that expression of the miRNA is positively regulated by the second product in the cell; and,
(f) engineering a transgene to express in the cell a transcript having a coding region encoding the first product and a 3′-non-coding region comprising one or more binding sites for the miRNA.

3. The method of claim 1 or 2, wherein the first product is a protein.

4. The method of claim 3, wherein the protein is MeCP2, optionally MeCP2 isoform e1 or MeCP isoform e2.

5. The method of any one of claims 2 to 4, wherein the second product is a protein, or nucleic acid, optionally wherein the nucleic acid is an miRNA.

6. The method of any one of claims 1 to 5, wherein the miRNA is miR-132.

7. The method of any one of claims 1 to 6, wherein the method further comprises engineering the 3′-non-coding region of the transcript to comprise one or more binding sites for one or more de-targeting miRNAs.

8. The method of claim 7, wherein the one or more de-targeting miRNAs inhibit expression of the transgene from liver, heart, lung, muscle, pancreas, or antigen presenting cells.

9. The method of claim 7 or 8, wherein the one or more de-targeting miRNA is miR-122, miR-1, or miR-122 and miR-1.

10. The method of any one of claims 1 to 9, wherein the step of engineering the transgene comprises inserting the transgene into a vector.

11. The method of claim 10, wherein the vector is a cloning vector, expression vector, plasmid, or viral vector.

12. A method of engineering a transgene, the method comprising:

(a) selecting a first gene encoding a first product in a cell;
(b) selecting an miRNA, the expression of which is positively regulated by the first product in the cell; and,
(c) engineering a transgene that expresses a transcript having a coding region encoding the first product and having one or more binding sites for the miRNA.

13. A method of engineering a transgene, the method comprising;

(a) selecting a first gene encoding a first product in a cell;
(b) selecting a second gene encoding a second product in the cell;
(c) determining that expression of the second product is positively regulated by the first product in the cell;
(d) selecting an miRNA;
(e) determining that expression of the miRNA is positively regulated by the second product in the cell; and,
(f) engineering a transgene to express in the cell a transcript having a coding region encoding the first product and having one or more binding sites for the miRNA.

14. A recombinant AAV (rAAV) vector for self-regulated expression of a protein, the rAAV vector comprising a nucleic acid engineered to express in a cell of a target tissue a transcript encoding the protein, wherein the transcript comprises at least one first miRNA binding site specific for a first miRNA, wherein expression of the first miRNA is positively regulated by expression of the protein in the cell.

15. A recombinant AAV comprising a capsid harboring the rAAV vector of claim 14, wherein the capsid comprises a capsid protein that facilitates selective transduction of the cell of the target tissue.

16. A recombinant nucleic acid encoding a transcript having i) a coding region encoding a protein and ii) two or more miRNA binding sites, wherein the two or more miRNA binding sites comprise:

(a) at least one first miRNA binding site specific for a first miRNA that is positively regulated by expression of the protein in a cell of a target tissue; and
(b) at least one second miRNA binding site specific for a second miRNA that is expressed, independent of expression of the protein, in cells of a non-target tissue.

17. A recombinant nucleic acid encoding a transcript having a coding region encoding human MeCP2 protein or a functional fragment thereof and a 3′-non-coding region comprising two or more miRNA binding sites, wherein the two or more miRNA binding sites comprise:

(a) at least one miRNA binding site specific for an miRNA that negatively regulates expression of the transcript; and
(b) at least one miRNA binding site specific for an miRNA that inhibits expression of the transcript in a cell of a non-target tissue.

18. The recombinant nucleic acid of claim 17, wherein the coding region encodes MeCP2 isoform e1.

19. The recombinant nucleic acid of claim 17 or 18, wherein the human MeCP2 comprises the sequence set forth in SEQ ID NO: 1.

20. The recombinant nucleic acid of any one of claims 17 to 19, wherein the at least one miRNA binding site specific for an miRNA that negatively regulates expression of the transcript comprises a miR-132 binding site, optionally wherein the at least one miRNA binding site is two or three miR-132 binding sites.

21. The recombinant nucleic acid of any one of claims 17 to 20, wherein the at least one miRNA binding site specific for an miRNA that inhibits expression of the transcript in a non-target tissue comprises a miR-1 binding site, mir-122 binding site, or miR-1 and miR-122 binding site.

22. The recombinant nucleic acid of any one of claims 17 to 21, wherein each of the one or more miRNA binding sites is located between the last codon of the coding region and the poly-A tail of the transcript.

23. The recombinant nucleic acid of any one of claims 17 to 22, further comprising a promoter, optionally a mouse MeCP2 promoter.

24. The recombinant nucleic acid of claim 23, wherein the mouse MeCP2 promoter comprises the sequence set forth in SEQ ID NO: 3.

25. The recombinant nucleic acid of any one of claims 17 to 24, wherein the recombinant nucleic acid is located on a plasmid.

26. A viral vector comprising the recombinant nucleic acid of any one of claims 17 to 24, optionally wherein the viral vector is an adeno-associated virus (AAV) vector, an adenovirus vector, a lentiviral vector, a herpesvirus vector, or a baculovirus vector.

27. A recombinant nucleic acid encoding a transcript having

(a) a coding region encoding human MeCP2 or a functional fragment thereof; and,
(b) a 3′-non-coding region comprising one or more miRNA binding sites, wherein transcript is flanked by adeno-associated virus (AAV) inverted terminal repeats (ITRs).

28. The recombinant nucleic acid of claim 27, wherein the coding region encodes MeCP2 isoform e1.

29. The recombinant nucleic acid of claim 27 or 28, wherein the human MeCP2 comprises the sequence set forth in SEQ ID NO: 1.

30. The recombinant nucleic acid of any one of claims 27 to 29, wherein the one or more miRNA binding sites are miR-1, miR-122, or miR-132 binding sites, or any combination thereof.

31. The recombinant nucleic acid of claim 30, wherein the transcript comprises a miR-1 binding site, a mir-122 binding site, and at least one miR-132 binding site.

32. The recombinant nucleic acid of claim 31, wherein the at least one miR-132 binding site is two or three mir-132 binding sites.

33. The recombinant nucleic acid of any one of claims 27 to 32, wherein the one or more miRNA binding sites are located between the last codon of the coding region and the poly-A tail of the transcript.

34. The recombinant nucleic acid of any one of claims 27 to 33, further comprising a promoter, optionally a mouse MeCP2 promoter.

35. The recombinant nucleic acid of claim 34, wherein the mouse MeCP2 promoter comprises the sequence set forth in SEQ ID NO: 3.

36. The recombinant nucleic acid of any one of claims 27 to 35, wherein the ITRs are AAV2 ITRs.

37. A recombinant adeno-associated virus (rAAV) comprising:

a capsid harboring the recombinant nucleic acid of any one of claims 17 to 24, or 27 to 36.

38. The rAAV of claim 37, wherein the nucleic acid comprises at least one ITR selected from an AAV2, AAV3, AAV4, AAV5, or AAV6 ITR.

39. The rAAV of claim 37 or 38, wherein the capsid comprises a capsid protein that facilitates passage of the rAAV across the blood-brain barrier.

40. The rAAV of claim 39, wherein the capsid protein has a serotype selected from the group consisting of AAV-PHP.B, AAV1, AAV2, AAV2i8, AAV2.5, AAV5, AAV6, AAV8, AAVrh8, AAV9, AAVrh10, AAV-B1, AAV9.45A-String (e.g., AAV9.45-AS), AAV9.45Angiopep, AAV9.47-Angiopep, AAV9.47-AS, AAV-CAM130, and AAV9HR.

41. The rAAV of any one of claims 37 to 40, wherein the capsid protein comprises or consists of a sequence set forth in SEQ ID NO: 14 or 15 (AAV-PHP.B, AAV9).

42. A composition comprising the recombinant nucleic acid of any one of claims 17 to 24, or 27 to 36, or the rAAV of any one of claims 37 to 41, and a pharmaceutically acceptable excipient.

43. The composition of claim 42, wherein the composition is formulated for injection, optionally wherein the injection is systemic injection (e.g., intravenous injection) or intrathecal injection.

44. A method of treating Rett syndrome in a subject, the method comprising, administering to a subject having or suspected of having Rett syndrome an effective amount of:

(a) the recombinant nucleic acid of any one of claims 17 to 24, or 27 to 36;
(b) the rAAV of any one of claims 37 to 41; or,
(c) the composition of claim 42 or 43.

45. The method of claim 44, wherein the subject is a human subject, optionally wherein the subject is less than one year old.

46. The method of claim 44 or 45, wherein the subject is characterized by a mutation in at least one copy of the MeCP2 gene, optionally wherein the mutation is a loss of function mutation.

47. The method of any one of claims 44 to 46, wherein the administration is injection, optionally systemic injection (e.g., intravenous injection) or intrathecal injection.

48. The method of any one of claims 44 to 47, wherein the administration results in the effective amount of (a), (b), or (c) crossing the blood-brain barrier of the subject.

49. The method of any one of claims 44 to 48, wherein the administration results in a non-toxic level of MeCP2 expression in the brain of the subject.

Patent History
Publication number: 20230416779
Type: Application
Filed: May 3, 2023
Publication Date: Dec 28, 2023
Applicant: University of Massachusetts (Boston, MA)
Inventors: Miguel Sena Esteves (Worcester, MA), Guangping Gao (Worcester, MA), Michael R. Green (Worcester, MA), Dan Wang (Worcester, MA), Tessa Mercedes Simone (Worcester, MA)
Application Number: 18/311,367
Classifications
International Classification: C12N 15/86 (20060101); C07K 14/47 (20060101);