US20190024121A1 - Tissue selective transgene expression - Google Patents

Tissue selective transgene expression Download PDF

Info

Publication number
US20190024121A1
US20190024121A1 US16/153,443 US201816153443A US2019024121A1 US 20190024121 A1 US20190024121 A1 US 20190024121A1 US 201816153443 A US201816153443 A US 201816153443A US 2019024121 A1 US2019024121 A1 US 2019024121A1
Authority
US
United States
Prior art keywords
cases
transgene
expression
viral vector
selective
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/153,443
Other languages
English (en)
Inventor
Stephanie TAGLIATELA
Andrew Young
Szu-Ying Chen
Kartik Ramamoorthi
David OBERKOFLER
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Encoded Therapeutics Inc
Original Assignee
Encoded Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Encoded Therapeutics Inc filed Critical Encoded Therapeutics Inc
Priority to US16/153,443 priority Critical patent/US20190024121A1/en
Publication of US20190024121A1 publication Critical patent/US20190024121A1/en
Assigned to ENCODED GENOMICS, INC. reassignment ENCODED GENOMICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, SZU-YING, OBERKOFLER, David, RAMAMOORTHI, Kartik, TAGLIATELA, Stephanie, YOUNG, ANDREW
Assigned to ENCODED THERAPEUTICS, INC. reassignment ENCODED THERAPEUTICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ENCODED GENOMICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/864Parvoviral vectors, e.g. parvovirus, densovirus
    • C12N15/8645Adeno-associated virus
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • A01K2217/052Animals comprising random inserted nucleic acids (transgenic) inducing gain of function
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/15Animals comprising multiple alterations of the genome, by transgenesis or homologous recombination, e.g. obtained by cross-breeding
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0312Animal model for Alzheimer's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14311Parvovirus, e.g. minute virus of mice
    • C12N2750/14341Use of virus, viral particle or viral elements as a vector
    • C12N2750/14343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/007Vectors comprising a special translation-regulating system cell or tissue specific

Definitions

  • Gene therapy has long been recognized for its enormous potential in how we approach and treat human diseases. Instead of relying on drugs or surgery, patients, especially those with underlying genetic factors, can be treated by directly targeting the underlying cause. Furthermore, by targeting the underlying genetic cause, gene therapy has the potential to effectively cure patients or provide sustained treatment over a longer period of time. Yet, despite this, clinical applications of gene therapy still require improvement in several aspects.
  • One area of concern is off target effects.
  • An attractive approach to address off target effects is to target gene expression of gene therapy to cell type(s) or tissue(s) of interest, or the target cell type(s) or tissue(s). As such, there is a need to identify elements and methods of use thereof for targeting gene therapy or gene expression to a tissue or cell type of interest.
  • compositions and methods for selective expression of a transgene in a target tissue or cell type over one or more non-target tissue or cell types comprise one or more regulatory elements (REs) which, when operably linked to a transgene (e.g., an ion channel subunit or a neurotransmitter regulator, or a syntaxin-binding protein), can facilitate or result in selective or preferential expression of the transgene in a target tissue or cell type (e.g., parvalbumin (PV) neurons) as compared to one or more non-target cell types (e.g., non-PV cells).
  • the REs are non-naturally occurring sequences.
  • the REs are human-derived regulatory elements.
  • the REs comprise a sequence from a non-human species, such as a monkey, or a dog, or a rabbit, or a mouse.
  • the compositions described herein are delivered into a cell in vivo, ex vivo, or in vitro using a viral vector and/or virus particles, such as adeno-associated virus (AAV) or lentivirus.
  • AAV adeno-associated virus
  • the compositions described herein are delivered into a cell as gene therapy.
  • methods and compositions for treating a neurological condition or disorder associated with a genetic defect in the CNS is a PV cell.
  • the neurological condition or disease is Dravet syndrome, Alzheimer's disease, epilepsy, and/or seizures.
  • the neurological condition or disease is a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • the present disclosure contemplates a nucleic acid cassette comprising one or more regulatory elements operably linked to a transgene that results in selective expression in any target cell type, e.g., PV neurons in the CNS, over one or more non-target cell types, or non-PV cells in the CNS.
  • each regulatory element comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • sequence identity is determined using BLAST.
  • at least one of the regulatory elements is human derived.
  • at least one of the regulatory elements is derived from a non-human mammal. In some cases, the regulatory elements are non-naturally occurring.
  • the regulatory elements result in selective expression of the transgene in PV neurons that is greater than expression of the same transgene when operably linked to a non-selective regulatory element, as measured by a co-localization assay.
  • the non-selective regulatory element is a constitutive promoter.
  • the non-selective regulatory element is any one of CAG, EF la, SV40, CMV, UBC, PGK, and CBA.
  • the regulatory elements result in selective expression of the transgene in PV neurons at a level that is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, or at least 20 fold as compared to selective expression of the transgene in PV neurons when operably linked to a non-selective regulatory element, as measured by the co-localization assay.
  • the regulatory elements result in selective expression in PV neurons that is at least 2%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% higher than expression in PV neurons when the transgene is operably linked to a non-selective regulatory element.
  • the regulatory elements result in selective expression in PV neurons that is about 1.5 times, 2 times, 2.5 times, 3 times, 3.5 times, 4 times, 4.5 times, 5 times, 5.5 times, 6 times, 6.5 times, 7 times, 7.5 times, 8 times, 8.5 times, 9 times, 9.5 times, 10 times, 15 times, 20 times, 25 times, 30 times, 40 times, 50 times, or 100 times higher than expected for natural distribution of PV neurons in the CNS.
  • the co-localization assay is an immunohistochemical assay.
  • the immunohistochemical assay comprises an anti-PV antibody.
  • the co-localization assay is performed as shown in Example 5 below.
  • the transgene encodes an ion channel subunit, a neurotransmitter regulator, a DNA binding domain, a gene editing protein, or a variant or a functional fragment thereof.
  • the ion channel subunit is an alpha subunit or a beta subunit of a sodium ion channel or a subunit of a potassium ion channel.
  • the transgene comprises any one of (i) SEQ ID NOs: 37-43; (ii) a functional fragment thereof; or (iii) a sequence having at least 80% sequence identity to (i) or (ii). In some cases, sequence identity is determined using BLAST.
  • the transgene comprises (i) SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, or KV3.3; (ii) a functional fragment thereof; or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the transgene is a neurotransmitter regulator that comprises (i) STXBP1, (ii) a functional fragment thereof, or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the transgene comprises a DNA binding protein that modulates expression of an endogenous gene.
  • the endogenous gene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, or STXBP1.
  • the transgene comprises a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • dCas nuclease-deactivated Cas
  • a deactivated transcription activator-like effector nuclease or a nuclease-deactivated zinc finger protein.
  • the transgene comprises a DNA binding domain linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the gene editing protein is a Cas protein.
  • the regulatory elements combined are less than 2.5 kb, less than 2 kb, less than 1.5 kb, less than 1 kb, or less than 500 bp in size.
  • the non-PV cells comprise one or more of non-PV cell types in the CNS.
  • the non-PV cells comprise one or more of excitatory neurons, dopaminergic neurons, astrocytes, microglia, and motor neurons.
  • the nucleic acid cassette is a linear construct.
  • the nucleic acid cassette is a vector.
  • the vector is a plasmid.
  • the vector is a viral vector.
  • the viral vector is an adeno-associated virus (AAV) vector.
  • AAV vector is AAV1, AAV8, AAV9, scAAV1, scAAV8, or scAAV9.
  • the viral vector is a lentiviral vector.
  • regulatory elements of any of the nucleic acid cassettes disclosed herein contain less than 600 bp of contiguous sequence from within 10 kb of the transcription start site of GAD2, GAD1, SYN1, NKX2.1, DLX1, DLXS/6, SST, PV, and/or VIP.
  • a method of treating a neurological disorder or condition in a subject in need thereof comprises delivering a therapeutically effective amount of any of the nucleic acid cassette disclosed herein.
  • the neurological disorder or condition is a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological disorder or condition is Dravet syndrome or Alzheimer's disease.
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • a method of increasing selective expression of a transgene in PV neurons in CNS comprises contacting a cell with a nucleic acid cassette disclosed herein.
  • the present disclosure contemplates a method of targeting expression of any transgene to PV neurons in the CNS, the method comprising operably linking one or more of PV neuron selective regulatory elements to a transgene.
  • each of the regulatory elements comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • sequence identity is determined using BLAST.
  • the regulatory elements result in selective expression of the transgene in PV neurons that is greater than expression of the same transgene when operably linked to a non-selective regulatory element, as measured by a co-localization assay.
  • the immunohistochemical assay comprises an anti-PV antibody (e.g., as described in Example 5 below).
  • the non-selective regulatory element is a constitutive promoter.
  • the non-selective regulatory element is any one of CAG, EF1 ⁇ , SV40, CMV, UBC, PGK, and CBA.
  • the regulatory elements result in selective expression of the transgene in PV neurons at a level that is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, or at least 20 fold as compared to a non-selective regulatory element when operably linked to the transgene, as measured by a co-localization assay.
  • the regulatory elements result in selective expression in PV neurons that is at least 2%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% higher than expression in PV neurons when the transgene is operably linked to a non-selective regulatory element.
  • the regulatory elements result in selective expression in PV neurons that is about 1.5 times, 2 times, 2.5 times, 3 times, 3.5 times, 4 times, 4.5 times, 5 times, 5.5 times, 6 times, 6.5 times, 7 times, 7.5 times, 8 times, 8.5 times, 9 times, 9.5 times, 10 times, 15 times, 20 times, 25 times, 30 times, 40 times, 50 times, or 100 times higher than expected for natural distribution of PV neurons in CNS.
  • the transgene is any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, a DNA binding protein, a gene editing protein, or a functional fragment thereof.
  • the regulatory elements and the transgene are in an AAV.
  • the AAV is AAV1, AAV8, AAV9, scAAV1, scAAV8, or scAAV9.
  • the present disclosure contemplates a method of treating a neurological condition or disorder in a subject in need thereof, the method comprising contacting a cell with a nucleic acid cassette comprising: one or more regulatory elements operably linked to a transgene that result in selective expression of the transgene in PV neurons over one or more non-PV cells in CNS.
  • each of the regulatory elements comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • sequence identity is determined using BLAST.
  • the transgene is a voltage-gated ion channel subunit, or a variant or a functional fragment thereof.
  • the subunit is a beta subunit of a sodium ion channel.
  • the subunit is an alpha subunit of a sodium ion channel. In some cases, the subunit is of a potassium ion channel.
  • the transgene is any one of (i) SCN1A, SCN1B, SCN2B, KV3.1, or KV3.3; (ii) a functional fragment thereof; or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the transgene is a DNA binding protein.
  • the DNA binding protein modulates an endogenous gene. In some cases, the endogenous gene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or STXBP1.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the transgene comprises a DNA binding domain linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene is a gene editing protein.
  • the gene editing protein is a Cas protein, e.g., Cas9.
  • the neurological condition or disorder is associated with a haploinsufficiency or a mutation in any of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or STXBP1.
  • the neurological condition or disorder is epilepsy, neurodegeneration, tauopathy, or neuronal hypoexcitability.
  • the neurological condition or disorder is Dravet syndrome.
  • the neurological condition or disorder is Alzheimer's disease.
  • the neurological condition or disease is a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • the regulatory elements of this disclosure result in selective expression of the transgene in PV neurons that is greater than expression of the same transgene when operably linked to a non-selective regulatory element, as measured by a co-localization assay.
  • the non-selective regulatory element is a constitutive promoter.
  • the non-selective regulatory element is any one of CAG, EF1 ⁇ , SV40, CMV, UBC, PGK, and CBA.
  • the regulatory elements result in selective expression in PV neurons at a level that is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, or at least 20 fold as compared to a non-selective regulatory element when operably linked to the transgene, as measured by a co-localization assay.
  • the regulatory elements result in selective expression in PV neurons that is at least 2%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% higher than expression in PV neurons when the transgene is operably linked to a non-selective regulatory element.
  • the regulatory elements result in selective expression in PV neurons that is about 1.5 times, 2 times, 2.5 times, 3 times, 3.5 times, 4 times, 4.5 times, 5 times, 5.5 times, 6 times, 6.5 times, 7 times, 7.5 times, 8 times, 8.5 times, 9 times, 9.5 times, 10 times, 15 times, 20 times, 25 times, 30 times, 40 times, 50 times, or 100 times higher than expected for natural distribution of PV neurons in CNS.
  • the nucleic acid cassette is in an AAV.
  • the AAV is AAV1, AAV8, AAV9, scAAV1, scAAV8, or scAAV9.
  • the present disclosure provides a method of treating Dravet syndrome, comprising contacting a cell with an AAV comprising a transgene, wherein the transgene is any one of (i) SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, or a DNA binding protein, (ii) a functional fragment thereof, or (iii) a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii). In some cases, sequence identity is measured using BLAST.
  • the DNA binding protein modulates an endogenous gene.
  • the DNA binding protein is a transcriptional modulator.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • dCas nuclease-deactivated Cas
  • a deactivated transcription activator-like effector nuclease or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene comprises a gene editing protein, e.g., a Cas protein, Cas9.
  • the endogenous gene is SCN1A, SNC2A, SNC8A, SCN1B, or SCN2B.
  • the AAV further comprises one or more PV neuron selective regulatory elements or one or more regulatory elements disclosed herein operably linked to the transgene.
  • each of the regulatory elements independently comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • the present disclosure provides a method of treating Alzheimer's disease, comprising contacting a cell with an AAV comprising a transgene, wherein the transgene is any one of (i) SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, or a DNA binding protein; (ii) a functional fragment thereof; or (iii) a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • sequence identity is measured using BLAST.
  • the DNA binding protein modulates an endogenous gene.
  • the endogenous gene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or STXBP1.
  • the transgene is a DNA binding protein comprising a transcriptional modulator.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene comprises a gene editing protein, e.g., a Cas protein, Cas9.
  • the AAV further comprises one or more PV neuron selective regulatory elements or one or more regulatory elements disclosed herein operably linked to the transgene.
  • each of the regulatory elements independently comprises each of the regulatory elements independently comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to (i) or (ii).
  • FIG. 1 illustrates the frequency of seizures (seizures per 12 hr interval) in SCN1A heterozygous mice after treatment with a recombinant AAVDJ vector comprising either SCN1B or eGFP operably linked to a regulatory element comprising a sequence of SEQ ID NO: 32.
  • the graph illustrates the mean values at each day of recording with error bars representing the standard error of the mean.
  • FIG. 2 illustrates high gamma power (50-100 Hz) of different mice: wild-type control (WT), untreated transgenic APP/PS1 mice (APP/PS1), or transgenic APP/PS1 mice treated with rAAV comprising SCN1B operably linked to a regulatory element comprising a sequence of SEQ ID NO: 32 (APP/PS1+SCN1B).
  • WT wild-type control
  • APP/PS1 untreated transgenic APP/PS1 mice
  • rAAV comprising SCN1B operably linked to a regulatory element comprising a sequence of SEQ ID NO: 32 (APP/PS1+SCN1B).
  • FIG. 3A illustrates immunofluorescence co-localization assay of CNS cells from pups following neonatal systemic injections of AAV9 comprising eGFP transgene operably linked to a regulatory element comprising a sequence of SEQ ID NO: 1 or SEQ ID NO: 8.
  • AAV9 comprising eGFP transgene operably linked to CAG was used as a control.
  • Lower row images illustrate eGFP+ cells.
  • Middle row images illustrate PV+ cells, which were stained with an anti-PV antibody.
  • Top row images (merge) illustrate an overlay of PV+, eGFP+ fluorescence (with representative eGFP+ and PV+ cells which are shown as white or light grey cells indicated by arrowheads) and DAPI+.
  • FIG. 3B illustrates the quantification of immunofluorescence co-localization studies illustrated in FIG. 3A , wherein selective expression in PV cells is expressed as the percentage of eGFP+ cells that were also PV+ in comparison to the CAG control, as measured by the immunofluorescence co-localization assay.
  • FIG. 4A illustrates immunofluorescence co-localization assay of CNS cells from adult mice following systemic injections of AAV9 comprising eGFP transgene operably linked to a regulatory element comprising a sequence of SEQ ID NO: 1 or SEQ ID NO: 8.
  • AAV9 comprising eGFP transgene operably linked to EF1 ⁇ was used as a control.
  • Lower row images illustrate eGFP+ cells.
  • Middle row images illustrate PV+ cells, which were stained with an anti-PV antibody.
  • Top row images (merge) illustrate an overlay of PV+ eGFP+ fluorescence (with representative eGFP+ and PV+ cells, or the white or light grey cells, indicated by arrowheads) and DAPI+.
  • FIG. 4B illustrates the quantification of immunofluorescence co-localization studies illustrated in FIG. 4A , wherein selective expression in PV cells is expressed as the percentage of eGFP+ cells that were also PV+ in comparison to the EF la control, as measured by the immunofluorescence co-localization assay.
  • FIGS. 5A-5F illustrate immunofluorescence co-localization assay of CNS cells from adult mice following direct CNS injections of AAVDJ comprising eGFP transgene operably linked to a regulatory element comprising a sequence of SEQ ID NOs: 2-22.
  • Lower row images illustrate eGFP+ cells.
  • Middle row images illustrate PV cells that were stained with an anti-PV antibody.
  • Top row images illustrate an overlay of PV+, eGFP+ fluorescence (with representative eGFP+ and PV+ cells, or the white or light grey cells, indicated by arrowheads) and DAPI+.
  • FIG. 5A illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising one of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, and SEQ ID NO: 5 operably linked to eGFP.
  • FIG. 5B illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising one of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO: 9 operably linked to eGFP.
  • FIG. 5C illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising one of SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, and SEQ ID NO: 13 operably linked to eGFP.
  • FIG. 5D illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising one of SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17 operably linked to eGFP.
  • FIG. 5E illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising one of SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 operably linked to eGFP.
  • 5F illustrates the immunofluorescence co-localization assay performed with AAVDJ comprising SEQ ID NO: 22 or SEQ ID NO: 34 operably linked to eGFP, wherein SEQ ID NO: 34 is a previously characterized non-selective regulatory element and was used as a control for comparison.
  • FIG. 6 illustrates the quantification of immunofluorescence co-localization studies illustrated in FIGS. 5A-5F , wherein selective expression in PV cells is expressed as the percentage of eGFP+ cells that were also PV+ in comparison to SEQ ID NO: 34, as measured by the immunofluorescence co-localization assay.
  • FIG. 7 illustrates a schematic of an example of an expression cassette containing REs of this disclosure, e.g., an enhancer, a promoter, and stability elements.
  • REs can be located upstream and/or downstream of a transgene in an expression cassette, which can be a plasmid, vector, or a viral vector.
  • compositions and methods of using such compositions in gene therapy to treat a disease or condition associated with the central nervous system e.g., Dravet syndrome, Alzheimer's disease, epilepsy, and/or seizures.
  • CNS central nervous system
  • Gene therapy can replace, modify, delete, or add a gene or a specific nucleic acid sequence, such as an expression cassette, to impart a therapeutic effect in a cell.
  • gene therapy is used to deliver an expression cassette into a cell that produces or results in a therapeutic effect.
  • a virus such as AAV, comprising a viral vector that comprises an expression cassette can be used to deliver a transgene into a cell.
  • the expression cassette can contain a transgene that provides a therapeutic effect when expressed in a cell.
  • One challenge in gene therapy is ensuring that the transgene is expressed in an appropriate cell type of interest, or the target cell type, to effect or target gene expression.
  • Traditional methods for targeting gene therapy have often relied on delivery methods and/or vehicles (e.g., varying the viruses used or capsid sequences of viruses).
  • delivery methods and/or vehicles e.g., varying the viruses used or capsid sequences of viruses.
  • another challenge in the field is increasing gene expression, especially when the gene is large, in a target cell type or tissue to exert a therapeutic effect.
  • the present disclosure provides a plurality of regulatory elements, which are non-coding nucleotide sequences, that can be operably linked to any transgene to increase or to improve selectivity of the transgene expression in the CNS, e.g., in PV neurons.
  • regulatory elements which are non-coding nucleotide sequences, that can be operably linked to any transgene to increase or to improve selectivity of the transgene expression in the CNS, e.g., in PV neurons.
  • one or more regulatory elements can be operably linked to any transgene in an expression cassette to modulate gene expression in a cell, such as targeting expression of the transgene in a target cell type or tissue (e.g., PV cells) over one or more non-target cell type or tissue (e.g., non-PV CNS cell-types).
  • targeting expression of the transgene in a target cell type or tissue includes increased gene expression in the target cell type or tissue.
  • One or more regulatory elements operably linked to a transgene can be part of an expression cassette, which can be a linear or a circular construct, a plasmid, a vector, a viral vector, e.g., a vector of an adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • Such expression cassette can be adapted for gene therapy or delivery into a subject (e.g., a human, a patient, or a mammal).
  • a subject e.g., a human, a patient, or a mammal.
  • operably linking one or more regulatory elements to a gene results in targeted expression of the gene in a target tissue or cell type in the CNS, such as a parvalbumin (PV) neuron.
  • a regulatory elements e.g., SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto
  • a gene therapy comprises one or more regulatory elements disclosed herein, wherein the regulatory elements are operably linked to a transgene and drive selective expression of the transgene in PV neurons.
  • selective expression of a gene in PV neurons is used to treat a disease or condition associated with a haploinsufficiency and/or a genetic defect in an endogenous gene, wherein the genetic defect can be a mutation in the gene or dysregulation of the gene.
  • the genetic defect can result in a reduced level of the gene product and/or a gene product with impaired function and/or activity.
  • an expression cassette comprises a gene, a subunit, a variant or a functional fragment thereof, wherein gene expression from the expression cassette is used to treat the disease or condition associated with the genetic defect, impaired function and/or activity, and/or dysregulation of the endogenous gene.
  • the disease or condition is Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizures.
  • the transgene is an ion channel or a neurotransmitter regulator, a DNA binding protein, or a subunit, variant, or functional fragment thereof.
  • the transgene is a sodium ion channel alpha subunit, sodium ion channel beta subunit, or a variant or functional fragment thereof.
  • the transgene is a potassium ion channel or a subunit thereof.
  • the transgene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein (e.g., a DNA binding protein that modulates expression of an endogenous gene), or a variant or functional fragment thereof.
  • the transgene comprises a sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein, or a variant or functional fragment thereof.
  • the transgene is a DNA binding protein that modulates expression of an endogenous gene, such as any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, and STXBP1.
  • determining can be used interchangeably herein to refer to any form of measurement, and include determining if an element is present or not (for example, detection). These terms can include both quantitative and/or qualitative determinations. Assessing may be relative or absolute.
  • expression refers to the process by which a nucleic acid sequence or a polynucleotide is transcribed from a DNA template (such as into mRNA or other RNA transcript) and/or the process by which a transcribed mRNA is subsequently translated into peptides, polypeptides, or proteins.
  • Transcripts and encoded polypeptides may be collectively referred to as “gene product.” If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell.
  • operably linked refers to juxtaposition of genetic elements, e.g., a promoter, an enhancer, a polyadenylation sequence, etc., wherein the elements are in a relationship permitting them to operate in the expected manner.
  • a regulatory element which can comprise promoter and/or enhancer sequences, is operatively linked to a coding region if the regulatory element helps initiate transcription of the coding sequence. There may be intervening residues between the regulatory element and coding region so long as this functional relationship is maintained.
  • a “vector” as used herein refers to a macromolecule or association of macromolecules that comprises or associates with a polynucleotide and which can be used to mediate delivery of the polynucleotide to a cell.
  • vectors include plasmids, viral vectors, liposomes, and other gene delivery vehicles.
  • the vector generally comprises genetic elements, e.g., regulatory elements, operatively linked to a gene to facilitate expression of the gene in a target.
  • the combination of regulatory elements and a gene or genes to which they are operably linked for expression is referred to as an “expression cassette”.
  • AAV is an abbreviation for adeno-associated virus, and may be used to refer to the virus itself or a derivative thereof. The term covers all serotypes, subtypes, and both naturally occurring and recombinant forms, except where required otherwise.
  • rAAV refers to recombinant adeno-associated virus, also referred to as a recombinant AAV vector (or “rAAV vector”).
  • AAV includes AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, rh10, and hybrids thereof, avian AAV, bovine AAV, canine AAV, equine AAV, primate AAV, non-primate AAV, and ovine AAV.
  • TRs native terminal repeats
  • Rep proteins Rep proteins
  • capsid subunits are known in the art. Such sequences may be found in the literature or in public databases such as GenBank.
  • rAAV vector refers to an AAV vector comprising a polynucleotide sequence not of AAV origin (i.e., a polynucleotide heterologous to AAV), typically a sequence of interest for the genetic transformation of a cell.
  • the heterologous polynucleotide is flanked by at least one, and generally by two, AAV inverted terminal repeat sequences (ITRs).
  • ITRs AAV inverted terminal repeat sequences
  • the term rAAV vector encompasses both rAAV vector particles and rAAV vector plasmids.
  • An rAAV vector may either be single-stranded (ssAAV) or self-complementary (scAAV).
  • An “AAV virus” or “AAV viral particle” or “rAAV vector particle” refers to a viral particle composed of at least one AAV capsid protein and an encapsidated polynucleotide rAAV vector. If the particle comprises a heterologous polynucleotide (i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it is typically referred to as an “rAAV vector particle” or simply an “rAAV vector”. Thus, production of rAAV particle necessarily includes production of rAAV vector, as such a vector is contained within an rAAV particle.
  • a heterologous polynucleotide i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell
  • the terms “treat”, “treatment”, “therapy” and the like refer to obtaining a desired pharmacologic and/or physiologic effect, including, but not limited to, alleviating, delaying or slowing the progression, reducing the effects or symptoms, preventing onset, inhibiting, ameliorating the onset of a diseases or disorder, obtaining a beneficial or desired result with respect to a disease, disorder, or medical condition, such as a therapeutic benefit and/or a prophylactic benefit.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease or at risk of acquiring the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • a therapeutic benefit includes eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder.
  • the compositions are administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • the methods of the present disclosure may be used with any mammal.
  • the treatment can result in a decrease or cessation of symptoms (e.g., a reduction in the frequency or duration of seizures).
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • the term “effective amount” or “therapeutically effective amount” refers to that amount of a composition described herein that is sufficient to affect the intended application, including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in a target cell.
  • the specific dose will vary depending on the particular composition chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • a “fragment” of a nucleotide or peptide sequence is meant to refer to a sequence that is less than that believed to be the “full-length” sequence.
  • a “variant” of a molecule refers to allelic variations of such sequences, that is, a sequence substantially similar in structure and biological activity to either the entire molecule, or to a fragment thereof.
  • a “functional fragment” of a DNA or protein sequence possesses at least a biologically active fragment of the sequence, which refers to a fragment that retains a biological activity (either functional or structural) that is substantially similar to a biological activity of the full-length DNA or protein sequence.
  • a biological activity of a DNA sequence can be its ability to influence expression in a manner known to be attributed to the full-length sequence.
  • a functional fragment of a regulatory element will retain the ability to influence transcription as the full-length RE.
  • subject and “individual” are used interchangeably herein to refer to a vertebrate, preferably a mammal, more preferably a human. “Subject” refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in human therapeutics, veterinary applications, and/or preclinical studies in animal models of a disease or condition. In some case, the subject is a mammal, and in some cases, the subject is human.
  • in vivo refers to an event that takes place in a subject's body.
  • in vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject.
  • in vitro assays encompass cell-based assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • Sequence comparisons such as for the purpose of assessing identities, mutations, or where one or more positions of a test sequence fall relative to one or more specified positions of a reference sequence, may be performed by any suitable alignment algorithm, including but not limited to the Needleman-Wunsch algorithm (see, e.g., the EMBOSS Needle aligner available at www.ebi.ac.uk/Tools/psa/emboss needle/, optionally with default settings), the BLAST algorithm (see, e.g., the BLAST alignment tool available at blast.ncbi.nlm.nih.gov/Blast.cgi, optionally with default settings), and the Smith-Waterman algorithm (see, e.g., the EMBOSS Water aligner available at www.ebi.ac.uk/Tools/psa/emboss water/, optionally with default settings). Optimal alignment may be assessed using any suitable parameters of a chosen algorithm, including default parameters.
  • sequence identity or “sequence homology”, which can be used interchangeably, refer to an exact nucleotide-to-nucleotide or amino acid-to-amino acid correspondence of two polynucleotides or polypeptide sequences, respectively.
  • techniques for determining sequence identity include determining the nucleotide sequence of a polynucleotide and/or determining the amino acid sequence encoded thereby, and comparing these sequences to a second nucleotide or amino acid sequence. Two or more sequences (polynucleotide or amino acid) can be compared by determining their “percent identity”, also referred to as “percent homology”.
  • the percent identity to a reference sequence may be calculated as the number of exact matches between two optimally aligned sequences divided by the length of the reference sequence and multiplied by 100. Percent identity may also be determined, for example, by comparing sequence information using the advanced BLAST computer program, including version 2.2.9, available from the National Institutes of Health.
  • the BLAST program is based on the alignment method of Karlin and Altschul, Proc. Natl. Acad. Sci. USA 87:2264-2268 (1990) and as discussed in Altschul, et al., J. Mol. Biol.
  • the BLAST program defines identity as the number of identical aligned symbols (i.e., nucleotides or amino acids), divided by the total number of symbols in the shorter of the two sequences. The program may be used to determine percent identity over the entire length of the sequences being compared. Default parameters are provided to optimize searches with short query sequences, for example, with the blastp program.
  • the program also allows use of an SEG filter to mask-off segments of the query sequences as determined by the SEG program of Wootton and Federhen, Computers and Chemistry 17: 149-163 (1993). Ranges of desired degrees of sequence identity are approximately 80% to 100% and integer values there between. Typically, the percent identities between a disclosed sequence and a claimed sequence are at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%. In general, an exact match indicates 100% identity over the length of the reference sequence. In some cases, reference to percent sequence identity refers to sequence identity as measured using BLAST (Basic Local Alignment Search Tool). In other cases, ClustalW can be used for multiple sequence alignment.
  • BLAST Basic Local Alignment Search Tool
  • Regulatory elements are nucleic acid sequences or genetic elements which are capable of influencing (e.g., increasing or decreasing) expression of a gene and/or confer selective expression of a gene (e.g., a reporter gene such as eGFP, a transgene, or a therapeutic gene) in a particular tissue or cell type of interest.
  • a regulatory element can be a transgene, an intron, a promoter, an enhancer, UTR, insulator, a repressor, an inverted terminal repeat (ITR) sequence, a long terminal repeat sequence (LTR), stability element, posttranslational response element, or a polyA sequence, or a combination thereof.
  • the regulatory element is a promoter or an enhancer, or a combination thereof.
  • the regulatory element is derived from a human sequence.
  • the cell type of interest is a PV neuron.
  • Regulatory elements can function at the DNA and/or the RNA level. Regulatory elements can function to modulate gene expression selectivity in a cell type of interest. Regulatory elements can function to modulate gene expression at the transcriptional phase, post-transcriptional phase, or at the translational phase of gene expression. Regulatory elements include, but are not limited to, promoter, enhancer, repressor, silencer, and insulator sequences.
  • regulation can occur at the level of translation (e.g., stability elements that stabilize mRNA for translation), RNA cleavage, RNA splicing, and/or transcriptional termination.
  • regulatory elements can recruit transcriptional factors to a coding region that increase gene expression selectivity in a cell type of interest.
  • regulatory elements can increase the rate at which RNA transcripts are produced, increase the stability of RNA produced, and/or increase the rate of protein synthesis from RNA transcripts.
  • regulatory elements can prevent RNA degradation and/or increase its stability to facilitate protein synthesis.
  • regulatory elements suppress transcription and/or translation processes in off-target cell-types.
  • off-target cell-types include, but are not limited to, excitatory neurons, non-PV CNS cell-types, and non-neuronal CNS cell types.
  • Various assays including, but not limited to, DNAase hypersensitivity, ATAC-Seq, and ChIP-Seq can be used to identify putative non-coding regulatory elements (REs).
  • the enzymatic reaction in each of these assays preferentially targets open/accessible chromatin states, a state which is thought to be predictive of regulatory elements.
  • To discover cell-type selective regulatory elements one can assay for open chromatin sequence for target cell-type of interest (e.g., parvalbumin neurons) and compare that to open chromatin sequences for non-target cell types (e.g., excitatory neurons). Additional filters can be applied to further refine target selection, including proximity to a cell-type selective gene, species conservation, and/or sequence motifs, such as transcription factor binding sites.
  • DNA sequences that are uniquely identified in the target cell type can be synthesized and cloned into an expression vector.
  • the selectivity of a regulatory element can be determined using immunohistochemical methods to quantify co-localization to known cell-
  • one method of isolating a cell-type selective regulatory element includes isolating nuclei from a brain tissue or cell type of interest from an animal model, which can be achieved by using an affinity purification method that isolates the tissue or cell type of interest (e.g., using beads coated to an anti-PV antibody for isolating PV neurons), using high-throughput natural priming and DNA synthesis to generate a pool of sequences from open chromatin regions in the nuclei, sequencing the pool of sequences to identify putative sequences that drive gene expression in the tissue or cell type of interest, and verifying selective expression in a reporter system in a cell line in vitro and/or in an animal model.
  • an affinity purification method that isolates the tissue or cell type of interest (e.g., using beads coated to an anti-PV antibody for isolating PV neurons)
  • high-throughput natural priming and DNA synthesis to generate a pool of sequences from open chromatin regions in the nuclei
  • sequencing the pool of sequences to identify putative sequences
  • Another method for identifying candidate regulatory elements that are selective in a tissue or cell type of interest include using R26-CAG-LSL-Sun1-sfGFP-Myc knockin mouse for harvesting the tissue or cell type of interest, isolating GFP+/Myc+ nuclei from the mouse neocortex of this strain using affinity purification, e.g., using anti-GFP or anti-Myc antibodies and protein G-coated magnetic beads to isolate nuclei from the neocortex.
  • Nuclear RNA from purified nuclei or whole neocortical nuclei can be converted to cDNA and amplified with the Nugen Ovation RNA-seq System V2 (Nugen 7102), followed by sequencing using the Illumina HiSeq 2500.
  • Genomic DNA from purified nuclei can be fragmented and used to make MethylC-seq libraries, which can be sequenced using the Illumina HiSeq 2000.
  • nuclei bound to beads are transposed using Tn5 transposase (Illumina FC-121-1030). After 9-12 cycles of PCR amplification, libraries are sequenced using an Illumina HiSeq 2500.
  • excitatory neuron nuclei can be digested to mononucleosomes using micrococcal nuclease, followed by salt extraction of chromatin, and native ChIP and library construction, which can be sequenced on an Illumina HiSeq 2500. After sequencing these libraries, the sequences are mapped to identify, for example, correlation in cell-type-specific hypo-methylation in CG-rich regions, histone modifications, transcriptional factor binding sites, and patterns associated with highly expressed transcriptional factors. Overlapping features and correlations from multiple assays and/or libraries described above provide evidence for identifying candidate sequences within such genomic regions as potential regulatory elements associated with selective expression and/or high expression in the cells isolated from the neocortex.
  • genomic region characterized by a strong overlap between hypomethylation detected in the methylC-seq library, ChIP assay, and an enrichment in transcription factor binding motifs in the same region provide convergent data that indicate the genomic region contains a sequence of a putative regulatory element selective for the tissue or cell type isolated.
  • PV neuron selective regulatory elements one can isolate PV neurons and purify nuclei from the isolated PV cells so that genomic sequences that are identified as active in multiple sequencing assays described above have a high likelihood of being PV cell-selective regulatory elements, e.g., a genomic region that is identified as active in an ATAC-seq assay (corresponding to regions of open chromatin), active in RNA-seq (indicative of active gene expression and low DNA methylation patterns in the region), and active in methylC-seq assay (which generates single-base resolution methylome maps from a cell type of interest).
  • a genomic region that is identified as active in an ATAC-seq assay corresponding to regions of open chromatin
  • active in RNA-seq indicator of active gene expression and low DNA methylation patterns in the region
  • methylC-seq assay which generates single-base resolution methylome maps from a cell type of interest.
  • sequences within the region can be generated using PCR methods and tested in additional assays in vitro and/or in vivo to validate tissue or cell type selectivity of the sequences.
  • validation assays include immunohistochemical co-localization assay, wherein an antibody or any detectable marker is used to label the cell type of interest and a second detectable marker, e.g., a fluorescent transgene, is operably linked to the putative regulatory elements.
  • a second detectable marker e.g., a fluorescent transgene
  • Selective expression driven by one or more putative regulatory elements can be validated by measuring the overlap between the cell type of interest (as measured by the detectable signal or fluorescence from its labeled marker, e.g., an anti-PV antibody) and the second detectable marker corresponding to expression of the transgene (e.g., eGFP or RFP) operably linked to the regulatory elements.
  • the detectable signal or fluorescence from its labeled marker e.g., an anti-PV antibody
  • the second detectable marker corresponding to expression of the transgene e.g., eGFP or RFP
  • An overlap in the signals from both detectable markers indicates cell-type selectivity in the labeled cell type if the amount of overlap observed is higher than the overlap observed when the regulatory elements are replaced with a control, such as CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), a non-selective regulatory element, or a previously characterized non-selective regulatory element.
  • a control such as CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), a non-selective regulatory element, or a previously characterized non-selective regulatory element.
  • a number of mouse lines that express Cre in a particular cell type can be used because cell-type selective Cre expression can drive Cre-induced expression of a fluorescent protein, such as RFP, in a cell type of interest.
  • Labeling such cell type of interest in vivo allows one to determine level of cell-type selective expression that is associated with a putative regulatory element operably linked to a fluorescent or reporter transgene in the same mouse. Similar to the co-localization assay, an overlap of the signals from both markers that exceeds the overlap detected for CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element is indicative of cell type selectivity for the regulatory elements tested.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • the mouse strain used is B6 PV-Cre mouse (Jackson Laboratory), which is a B6 PV-Cre knock-in mouse that expresses Cre recombinase in parvalbumin-expressing neurons (e.g., interneurons in the brain and proprioceptive afferent sensory neurons in the dorsal root ganglia), without disrupting endogenous Pvalb expression.
  • B6 PV-Cre mouse Jackson Laboratory
  • Cre recombinase in parvalbumin-expressing neurons e.g., interneurons in the brain and proprioceptive afferent sensory neurons in the dorsal root ganglia
  • sequences of such regulatory elements can be varied using various mutagenesis methods, e.g., error-prone PCR methods, to improve its selectivity.
  • two or more regulatory elements having cell selectivity can be combined.
  • combined regulatory elements exhibit enhanced cell-type selectivity in driving gene expression in the cell type of interest.
  • such regulatory elements are truncated one or more bases at a time to determine the minimal amount of sequence that retains its cell type selectivity.
  • the present disclosure provides a plurality of nucleotide sequences that are regulatory elements.
  • any one or more of the regulatory elements disclosed herein result in increased selectivity in gene expression in a parvalbumin cell.
  • regulatory elements disclosed herein are PV-cell-selective.
  • PV cell selective regulatory elements are associated with selective gene expression in PV cells more than expression in non-PV CNS cell-types.
  • PV cell selective regulatory elements as associated with reduced gene expression in non-PV CNS cell types.
  • Non-limiting examples of regulatory elements include SEQ ID NOs: 1-32, as provided in TABLE 1 below.
  • regulatory elements disclosed herein are cell-type selective. In some cases, regulatory elements disclosed herein are selective for PV neurons. In some cases, regulatory elements disclosed herein are selective for PV neurons in the CNS. In some cases, PV-cell selective regulatory elements or any regulatory elements disclosed herein can result in selective gene expression in PV neurons over at least one, two, three, four, five, or more non-PV CNS cell-types.
  • any one or more of the regulatory elements disclosed herein are operably linked to a transgene in an expression cassette to result in selective expression in a target cell-type, e.g., a PV neuron.
  • a regulatory element of any of the embodiments herein comprises or consists of any one of (i) SEQ ID NOs: 1-33; (ii) a variant, functional fragment, or a combination thereof; or (iii) a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to any one of (i) or (ii).
  • a regulatory element comprises any one of SEQ ID NOs: 1-32.
  • sequence identity is measured by BLAST.
  • two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto, are combined to form a larger regulatory element, or are operably linked to a gene in an expression cassette.
  • two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto, are combined using a linker sequence of 1-50 nucleotides.
  • two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto, are combined without a linker sequence.
  • a sequence of SEQ ID NO: 33 is used as a linker between any two regulatory elements.
  • a linker sequence between any two regulatory elements comprises SEQ ID NO: 33 or a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 33.
  • sequence identity is measured by BLAST.
  • the regulatory elements need not be adjacent or linked in an expression cassette.
  • one regulatory element can be located upstream of a transgene, while a second regulatory element and/or additional regulatory elements can be located downstream of the transgene.
  • one or more regulatory elements can be located upstream of a transgene.
  • one or more regulatory elements can be located downstream of a transgene.
  • any one or more of SEQ ID NOs: 1-22, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto can be combined with any one or more of SEQ ID NO: 23-30, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto, to form a larger regulatory element.
  • a regulatory element comprises SEQ ID NO: 1 and SEQ ID NO: 30.
  • a regulatory element comprises SEQ ID NO: 8 and SEQ ID NO: 30.
  • a regulatory element comprises SEQ ID NO: 1 and SEQ ID NOs: 23-29.
  • a regulatory element comprises SEQ ID NO: 8 and SEQ ID NOs: 23-29.
  • a regulatory element comprises SEQ ID NO: 30 or any one or more of SEQ ID NOs: 23-29, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • one or more regulatory elements of the present disclosure result in selective gene expression in a PV cell.
  • regulatory elements that show selective activity or function in a target cell type also show minimal activity or function in one or more off-target cell-types, e.g., non-PV CNS cell-types, non-inhibitory neurons or excitatory neurons, non-PV cells.
  • one or more regulatory elements operably linked to a gene modulates gene expression in a cell, including but not limited to, selective expression in a target cell-type over non-target cell-types.
  • Selective expression in a target cell or cell type can also be referred to as cell-selective expression or cell-type selective expression.
  • Selective expression generally refers to expression in a high fraction of cells of the cell type of interest (or the target cell type) as compared to other cells (or non-target cell type). Selective expression can also be viewed as preferential expression in a target cell or target cell type over one or more non-target cells or cell-types.
  • selective expression of one or more regulatory elements of this disclosure is compared to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element that is known to drive expression in any cell or cell type without selectivity.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • selective expression of one or more regulatory elements of this disclosure is compared to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), a non-selective regulatory element, or an expression cassette without the regulatory elements.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • Non-target cell types can include a different subset, subtype, or type of cells as compared to the target cell or target cell type, or all non-target cell types.
  • one or more regulatory elements operably linked to a gene result in selective expression in a target cell type over at least one type of non-target cells, or at least two, at least three, at least four, at least five, or more than five types of non-target cells.
  • non-target cell types refer to all other cell types not including the target cell type.
  • non-target cell types are all other cell types within a relevant tissue or organ not including the target cell type, e.g., all non-target cell types in the CNS, all non-target cell types in the hippocampus.
  • non-target cell or non-target cell type encompasses a subset or subtype of cells that is not the target cell.
  • non-PV CNS cell-types can include GABAergic cells that express calretinin and/or somatostatin instead of parvalbumin, or all GABAergic cells that do not express parvalbumin.
  • cell types are distinguished by having a different cell marker, morphology, phenotype, genotype, function, and/or any other means for classifying cell types.
  • Selectivity of expression driven by a regulatory element in a cell or cell type of interest can be measured in a number of ways.
  • Selectivity of gene expression in a target cell type over non-target cell types can be measured by comparing the number of target cells that express a detectable level of a transcript from a gene that is operably linked to one or more regulatory elements to the total number of cells that express the gene.
  • Such measurement, detection, and quantification can be done either in vivo or in vitro.
  • selectivity for PV neurons can be determined using a co-localization assay.
  • the co-localization assay is based on immunohistochemistry.
  • a detectable reporter gene is used as a transgene to allow the detection and/or measurement of gene expression in a cell.
  • a detectable marker e.g., a fluorescent marker or an antibody, which specifically labels the target cell is used to detect and/or measure the target cells.
  • a co-localization assay employs imaging, e.g., fluorescent imaging, to determine the overlap between different fluorescent labels, e.g., overlap between a fluorescence signal indicative of a target cell and another fluorescence signal indicative of gene expression.
  • fluorescent labels used for a co-localization assay include a red fluorescent protein (RFP), such as a tdTomato reporter gene, and a green fluorescent reporter protein, such as eGFP.
  • RFP red fluorescent protein
  • eGFP green fluorescent reporter protein
  • a gene operably linked to one or more regulatory elements is a fluorescent protein, e.g., eGFP or RFP, wherein expression of the transgene provides a detectable signal.
  • tissue is stained for eGFP or fluorescence from eGFP is detected directly using a fluorescence microscope.
  • a second fluorescent marker or reporter gene having a different fluorescence or detectable signal can be used to indicate the target cells, such as an antibody that identifies the target cells.
  • an anti-PV antibody that interacts specifically with PV neurons can be used to yield a detectable signal that is distinguishable from the fluorescence used to measure gene expression, such as a red fluorescence or a red stain.
  • eGFP is a transgene operably linked to one or more regulatory elements that drive selective expression in PV neurons
  • the PV neurons are labeled with an anti-PV antibody
  • selectivity of gene expression in PV cells is measured as percentage of eGFP+ cells that are also PV+.
  • PV+ cells that are also eGFP+ are indicated by the overlap of both fluorescence signals, i.e., an overlap of the red and green fluorescence.
  • Such measurement, analysis, and/or detection can be done by eye inspection or by a computer.
  • selectivity of expression can also be measured by comparing the number of target cells that express a transgene operably linked to one or more regulatory elements to the total number of all cells that express the transgene. In both approaches, the higher the number of target cells that express the transgene, the more selective are the regulatory elements for the target cells.
  • the target cells are PV neurons.
  • one or more regulatory elements disclosed herein result in increased selectivity in gene expression in a PV neuron. In some cases, one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to non-PV CNS cell-types.
  • one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to non-PV GABAergic cells, wherein non-PV GABAergic cells can be any one or more of GABAergic cells that express calretinin (CR), somatostatin (SOM), cholecystokinin (CCK), neuropeptide Y (NPY), vasointestinal polypeptide (VIP), choline acetyltransferase (ChAT), or a combination thereof.
  • CR calretinin
  • SOM somatostatin
  • CCK cholecystokinin
  • NPY neuropeptide Y
  • VIP vasointestinal polypeptide
  • ChAT choline acetyltransferase
  • one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to at least one, at least two, at least three, at least four, at least five, or more than five non-PV GABAergic subtypes. In some cases, one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to all other non-PV GABAergic cells, or all other GABAergic cells that do not express PV, or all other CNS cells that do not express PV, or all other neurons that do not express PV. In some cases, one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to all non-PV cells in the CNS or all non-PV neurons.
  • one or more regulatory elements operably linked to a transgene result in selective expression of the transgene in PV cells, wherein the percentage of PV cells expressing the transgene is at a percentage higher than gene expression in PV cells wherein the transgene is operably linked to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element, such as SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80% sequence identity thereto.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • a non-selective regulatory element such as SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80% sequence identity thereto.
  • one or more regulatory elements result in selective expression in PV neurons at a level that is at least 1.5 fold, at least 2 hold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 15 fold, at least 20 fold, at least 25 fold, or at least 50 fold as compared to expression of a gene operably linked to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element such as SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • a non-selective regulatory element such as SEQ ID NO: 34, or
  • a regulatory element is human derived or comprises a sequence that is human derived.
  • a regulatory element is mouse derived or comprises a sequence that is mouse derived.
  • a regulatory element comprises a non-naturally occurring sequence.
  • a regulatory element is non-naturally occurring.
  • one or more human derived regulatory elements are combined with another regulatory element to generate a non-naturally occurring regulatory element.
  • a human derived regulatory element is combined with a mouse derived regulatory element.
  • human derived refers to sequences that are found in a human genome (or a human genome build), or sequences homologous thereto.
  • a homologous sequence may be a sequence which has a region with at least 80% sequence identity (e.g., as measured by BLAST) as compared to a region of the human genome. For example, a sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homologous to a human sequence is deemed human derived.
  • a regulatory element contains a human derived sequence and a non-human derived sequence such that overall the regulatory element has low sequence identity to the human genome, while a part of the regulatory element has 100% sequence identity (or local sequence identity) to a sequence in the human genome.
  • a human-derived regulatory element is a sequence that is 100% identical to a human sequence. In some instances, the sequence of a cell-type selective regulatory element is 100% human derived.
  • a regulatory element can have 50% of its sequence be human derived, and the remaining 50% be non-human derived (e.g., mouse derived or fully synthetic).
  • a regulatory element that is regarded as 50% human derived and comprises 300 bp may have an overall 45% sequence identity to a sequence in the human genome, while base pairs 1-150 of the RE may have 90% identity (local sequence identity) to a similarly sized region of the human genome.
  • a sequence that is homologous to a human derived regulatory sequence is at least 90% identical to a human sequence.
  • a regulatory element herein comprises a sequence that has at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to any one of SEQ ID NOs: 1, 23-31, and 33.
  • sequence identity is measured by BLAST.
  • a regulatory element comprises a sequence that is homologous (e.g., at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity) to any one or more of SEQ ID NOs: 23-29, or a functional fragment or combination thereof, such regulatory element results in higher expression of an operably linked transgene (when a promoter is also present in the expression vector or cassette), as compared to a similar vector without the regulatory element.
  • Such higher expression of a transgene can be observed, e.g., in HEK293T or CHO cells.
  • one or more regulatory elements comprise any one or more of SEQ ID NO: 23-29 combined with or used in combination with any one or more of SEQ ID NOs: 1-22 with or without a linker sequence such as SEQ ID NO: 33.
  • any two regulatory elements of this disclosure are linked together using a polynucleotide linker comprising 1-50 nucleotides, such as SEQ ID NO: 33 or a variant thereof.
  • the linker sequence is a human derived sequence.
  • the linker sequence is mouse derived or non-naturally occurring.
  • two regulatory elements are joined without a linker or without any intervening sequence.
  • a linker comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides.
  • the linker sequence is a result of restriction enzyme site, ligation, PCR, and/or cloning.
  • a human derived regulatory element is combined with a mouse derived regulatory element, such as SEQ ID NO: 32, which is a combination of SEQ ID NO: 8 (a mouse derived sequence) and SEQ ID NOs: 23-29 (human derived sequences).
  • cell-type selective regulatory elements are combined directly with no additional linker sequence.
  • cell-type selective regulatory elements are combined with one or more short linker sequences which can be either deliberate or cloning artifacts.
  • a linker sequence comprises 1-50 bases.
  • SEQ ID NO: 31 comprises the sequence of SEQ ID NOs: 1 and 23-29 along with an additional 19 bp of the genomic sequence (SEQ ID NO: 33) immediately following the sequence of SEQ ID NO: 1.
  • SEQ ID NO: 32 also includes these 19 bp but without the sequence of SEQ ID NO: 1.
  • the combined cell-type selective regulatory elements can include short sequences, generally less than 50 bp, less than 20 bp, less than 15 bp, or less than 10 bp, from a cloning plasmid or restriction enzyme recognition site.
  • regulatory elements can be derived from non-coding DNA sequences.
  • regulatory elements derived from non-coding DNA are associated with genes, such as upstream sequences, introns, 3′ and 5′ untranslated regions (UTRs), and/or downstream regions.
  • regulatory elements derived from non-coding DNA sequences are not associated with a gene.
  • regulatory elements are derived from coding sequences.
  • the genomic region from which a regulatory element is derived is distinct from the genomic region from which an operably linked transgene is derived.
  • a RE is derived from a distal genomic region or location with respect to the genomic region or location from which the transgene is derived (such as a naturally occurring or an endogenous version of the transgene).
  • a regulatory element is any non-coding sequence that modulates gene expression, e.g., selectivity of expression in a target cell.
  • the target cell is a PV neuron.
  • a regulatory element is derived from a genomic sequence upstream of a transcription initiation site, a 5′ UTR sequence, an exonic sequence, an intronic sequence, or a 3′ UTR sequence.
  • a human derived regulatory element comprises an intronic human derived sequence.
  • a regulatory element comprises an enhancer, and its presence in an expression cassette along with a promoter increases expression of an operably linked transgene in the target cell-type (e.g., PV neurons) as compared to expression of the same transgene by the promoter without the enhancer.
  • an enhancer increases expression of an operably linked transgene through either a transcriptional mechanism, posttranscriptional mechanism, or both.
  • a regulatory element comprises an enhancer sequence, a promoter sequence, or a combination of the enhancer and promoter sequences.
  • a regulatory element comprises one or more of a human derived enhancer sequence, a human derived promoter sequence, a human derived intronic sequence, and/or a combination thereof.
  • a regulatory element comprises one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto. In some cases, a regulatory element comprises one or more of SEQ ID NOs: 1-22, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a regulatory element comprises one or more of SEQ ID NOs: 23-29, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto. In some cases, a regulatory element comprises one or more of SEQ ID NOs: 30-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a regulatory element comprises a sequence of SEQ ID NOs: 30-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a regulatory element is derived from non-human DNA sequences or both human and non-human genomic sequences.
  • cell-type selective regulatory elements, or parts thereof are homologous to a mammalian genomic sequence.
  • the regulatory elements have at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to a mammalian genomic sequence.
  • a regulatory element is derived from a mouse genomic sequence.
  • a regulatory element, or fragments thereof has at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more than 99% identity to a mouse genomic sequence or a non-human mammalian genomic sequence.
  • sequence identity is measured by BLAST.
  • the regulatory elements may comprise any of SEQ ID NOs: 1-33, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • cell-type selective regulatory elements are short.
  • the size of the regulatory elements is compatible with the cloning capacity of a vector, e.g., a viral vector or rAAV, such that the combined size of a transgene and one or more regulatory elements does not exceed the cloning capacity of a vector.
  • the cell-type selective regulatory elements have a length of up to about 2050 bp, 2000 bp, 1900 bp, 1800 bp, 1700 bp, 1600 bp, 1500 bp, 1400 bp, 1300 bp, 1200 bp, 1100 bp, 1000 bp, 900 bp, 800 bp, 700 bp, 600 bp, 500 bp, 400 bp, 300 bp, 200 bp, or 100 bp.
  • the cell-type selective regulatory elements have a total length of no more than about 20 bp, 30 bp, 40 bp, 50 bp, 60 bp, 70 bp, 80 bp, 90 bp, 100 bp, 200 bp, 300 bp, 400 bp, 500 bp, 600 bp, 700 bp, 800 bp, 900 bp, 1000 bp, 1010 bp, 1020 bp, 1030 bp, 1040 bp, 1050 bp, 1060 bp, 1070 bp, 1080 bp, 1090 bp, 1100 bp, 1200 bp, 1300 bp, 1500 bp, 1600 bp, 1700 bp, 1800 bp, 1900 bp, or 2000 bp.
  • the cell-type selective regulatory elements have a length of about 100 bp-1100 bp, 100 bp-1000 bp, 100 bp-900 bp, 200 bp-900 bp, 200 bp-800 bp, 300 bp-600 bp, 400 bp-800 bp, 500 bp-600 bp, or 600 bp-900 bp.
  • a regulatory element is between about 400-600 bp, 400-600 bp, 400-700 bp, 400-800 bp, 400-900 bp, 400-1000 bp, or 400-1500 bp.
  • a regulatory element is between about 500-600 bp, 500-700 bp, 500-800 bp, 500-900 bp, 500-1000 bp, or 500-1500 bp. In some cases, two or more regulatory elements are combined to form a larger cell-type selective regulatory element of 1300-2500 bp, 1300-2060 bp, about 1350 bp, about 2050 bp, or about 1880 bp.
  • two or more cell-type selective regulatory elements can be combined.
  • two, three, four, five, six, seven, eight, nine, ten or more cell-type selective regulatory elements can be combined.
  • SEQ ID NO: 30 comprises sequences from seven regulatory elements, i.e., SEQ ID NOs: 23-29, all of which are derived from human genomic sequence.
  • cell-type selective regulatory elements refer to PV-neuron selective regulatory elements.
  • a cell-type selective regulatory element is repeated two or more times to make a combined regulatory element that is also cell-type selective or has enhance cell-type selective property.
  • two or more regulatory elements with different cell-type selectivity are combined.
  • a cell-type selective regulatory element is combined with a non-selective regulatory element, e.g., a non-selective enhancer element that drives high gene expression.
  • a promoter regulatory element with high selectivity for a target cell can be combined with a regulatory element with high efficiency of expression.
  • one or more cell type-selective regulatory elements are combined with one or more high efficiency regulatory elements.
  • any one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto can be combined with a constitutive promoter, such as a GAD2 promoter, a human synapsin promoter, a minCMV promoter, a TATA box, a super core promoter, or an EF1 ⁇ promoter, or a combination thereof.
  • a constitutive promoter such as a GAD2 promoter, a human synapsin promoter, a minCMV promoter, a TATA box, a super core promoter, or an EF1 ⁇ promoter, or a combination thereof.
  • the present disclosure provides a list of regulatory elements that can be added to any gene therapy to result in selective gene expression in a target cell type, such as a PV neuron over one or more non-target cell types, such as non-PV CNS cells, including but not limited to excitatory cells and/or non-PV GABAergic cells.
  • a target cell type such as a PV neuron over one or more non-target cell types, such as non-PV CNS cells, including but not limited to excitatory cells and/or non-PV GABAergic cells.
  • cell-type selective regulatory elements can be combined with other regulatory elements such as a high expressing promoter or a sequence that increases mRNA stability.
  • one or more cell-type selective regulatory elements are combined with a human, a non-human, or a non-mammalian sequence, for example a hSyn1 promoter, CBA promoter, a CMV promoter, an EF1 ⁇ promoter, a polyA signal (e.g., SV40 polyA signal), or a post-transcriptional regulatory element such as woodchuck hepatitis virus post-transcriptional regulatory element (WPRE).
  • WPRE woodchuck hepatitis virus post-transcriptional regulatory element
  • the combined regulatory elements can come from different species.
  • the combined regulatory elements can come from different genomic regions within a species.
  • regulatory elements are derived from distal genomic sequences, e.g., sequences that do not normally or naturally associate with each other or with a cell type of interest, are combined.
  • individual regulatory elements used to make a combined regulatory element can come from different human chromosomes.
  • a regulatory element of the disclosure comprises a functional fragment of any of SEQ ID NOs: 1-32, or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • Such functional fragment can increase expression of a transgene in an expression cassette or vector when compared to a similar expression cassette or vector without the regulatory element.
  • Such a functional fragment can function as an enhancer to increase cell-type selective expression when the fragment is operably linked to a transgene as compared to a similar vector or cassette without the functional fragment.
  • a fragment is preferably more than 30, 40, 50, or 60 bp in length.
  • a PV cell selective regulatory element or any regulatory element of this disclosure comprises any one of SEQ ID NOs: 1-32, (ii) a nucleic acid sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 1-32, (iii) a functional fragment of any sequence of (i) or (ii), or (iv) a combination of any sequence of (i), (ii) and/or (iii). In some cases, sequence identity is measured by BLAST.
  • two or more of SEQ ID NOs: 1-29, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto are used as a regulatory element to increase transgene expression selectively in PV cells as compared to non-PV CNS cells, or to in any target cell type as compared to non-target cell type.
  • a functional fragment is one that results in selective expression in a target cell type over one or more non-target cell types.
  • two or more copies of a regulatory element can be used to enhance selective expression in a target cell, e.g., two or more copies of any one of SEQ ID NOs: 1-29, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto are operably linked to another regulatory element, such as a promoter or enhancer, to further increase selective expression in a target cell.
  • another regulatory element such as a promoter or enhancer
  • one can enhance any gene therapy by adding one or more regulatory elements as disclosed herein to improve or increase expression from the gene therapy in a target cell as compared to non-target cells.
  • the target cell is PV neurons or GABAergic cells that express parvalbumin.
  • one or more regulatory elements e.g., any one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto
  • a regulatory element drives selective expression or preferential expression in a target cell subtype over at least one, at least two, at least three, at least four, at least five, or more than five non-target subtypes, or all other known subtypes of the cell.
  • GABAergic cells comprise different subtypes, including PV cells.
  • the target cell type is a PV cell.
  • one or more regulatory elements are selective for PV cells over at least one, at least two, at least three, at least four, at least five, or more than five non-target cell types. In some instances, one or more regulatory elements are selective for PV cells over all other known CNS cell-types.
  • any one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof. or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto can be combined with any one or more of SEQ ID NOs: 1-32, or a fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • such combined regulatory elements are linked using a linker of 1-50 nucleotides. In some cases, such combined regulatory elements are not linked.
  • one or more regulatory elements disclosed herein when operably linked to any transgene (e.g., a reporter transgene or a therapeutic transgene), drives selective expression or preferential expression in at least one target cell type at a level that is statistically significantly higher than the expression driven by CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element (e.g., SEQ ID NO: 34, or a fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto) when operably linked to the same transgene, or by the same construct without the regulatory elements.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • a non-selective regulatory element e.g., SEQ ID NO: 34, or a fragment thereof, or a
  • the regulatory elements drive selective expression in the target cell type at a level that is at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 times the expression level by CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element when operably linked to the same transgene, or by the same construct without the regulatory elements.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • such cell-type selective expression is assayed using a co-localization assay as described herein.
  • the target cell type is a parvalbumin cell.
  • such co-localization assay is conducted using an anti-PV antibody.
  • such co-localization assay is conducted using a PV-Cre mouse as disclosed herein.
  • the non-selective regulatory element is SEQ ID NO: 34, or a fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • an expression cassette and “nucleic acid cassette” are used interchangeably to refer to a polynucleotide molecule or a nucleic acid sequence.
  • an expression cassette comprises one or more regulatory elements disclosed herein operably linked to a transgene.
  • an expression cassette comprises one or more regulatory elements.
  • an expression cassette comprises one or more cell type selective regulatory elements disclosed herein.
  • an expression cassette comprises one or more PV cell selective regulatory elements disclosed herein.
  • the expression cassette further comprises a promoter.
  • an expression cassette comprises one or more sequences of SEQ ID NOs: 1-32 and/or any combination thereof.
  • an expression cassette comprises one or more of SEQ ID NOs: 1-32, (ii) a nucleic acid sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 1-32, (iii) a functional fragment of any sequence of (i) or (ii), or (iv) a combination of any sequence of (i), (ii) and/or (iii). In some cases, sequence identity is measured by BLAST.
  • a regulatory element is located upstream of a transgene in an expression cassette. In some cases, a regulatory element is located downstream of a transgene in an expression cassette. In some cases, an expression cassette further comprises a promoter, e.g., a hSyn1 promoter, CBA promoter, a CMV promoter, an EF1 ⁇ promoter, a polyA signal (e.g., SV40 polyA signal), or a post-transcriptional regulatory element such as woodchuck hepatitis virus post-transcriptional regulatory element (WPRE).
  • a promoter e.g., a hSyn1 promoter, CBA promoter, a CMV promoter, an EF1 ⁇ promoter, a polyA signal (e.g., SV40 polyA signal), or a post-transcriptional regulatory element such as woodchuck hepatitis virus post-transcriptional regulatory element (WPRE).
  • WPRE woodchuck hepatitis virus post-transcriptional regulatory
  • one or more regulatory elements described herein are operably linked to a transgene in an expression cassette.
  • a gene therapy comprises an expression cassette comprising a transgene operably linked to one or more, two or more, three or more, four or more, or five or more regulatory elements of the present disclosure to result in selective expression of the transgene in a target tissue or cell type, such as PV neurons.
  • an expression cassette comprises one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein operably linked to a transgene, e.g., a reporter gene, eGFP, SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein, or a variant or a fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a transgene e.g., a reporter gene, eGFP, SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein, or a variant or a fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • an expression cassette is adapted for delivery via gene therapy.
  • an expression cassette is a linear or a circular construct.
  • an expression cassette is part of a plasmid, vector, a viral vector, or rAAV.
  • a gene therapy is administered directly to the CNS of a subject in need thereof or systematically via injection and/or infusion.
  • such subject has been diagnosed with a disease or condition associated with a haploinsufficiency or a genetic mutation, such as a haploinsufficiency or a mutation in any one of the following genes: SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, or STXBP1.
  • the subject is at risk for or has Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizures.
  • such gene therapy is delivered using a virus or a viral vector, such as rAAV.
  • a virus or a viral vector such as rAAV.
  • an AAV serotype with a tropism for CNS cells and/or ability to cross the blood brain barrier is used, such as AAV9 or a variant thereof.
  • one or more regulatory elements operably linked to a transgene in an expression cassette result in selective gene expression in PV cells as compared to non-PV CNS cells, or as compared to a control element, such as a CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto).
  • a control element such as a CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at
  • regulatory elements result in at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of all cells expressing the transgene are PV neurons. In some cases, regulatory elements result in selective gene expression in PV neurons that is about 1.5 times, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 7.5 times, 8 times, 9 times, or 10 times higher than expected for natural distribution of PV neurons in CNS.
  • a regulatory element drives selective expression in PV cells, wherein the percentage of PV cells expressing the transgene is at a percentage that is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, or at least 10 fold higher than the expected distribution of PV cells in the CNS, or at least 1-5%, 5%-10%, 10-15%, 15-20%, 20-25%, 25-30%, 30-35%, 35-40%, 40-45%, 45-50%, 50-55%, 55-60%, 65-70%, 70-75%, 75-80%, 80-85%, 85-90%, or 90-95% higher than the expression in PV cells when the transgene is operably linked to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element having a sequence of SEQ ID NO: 34, or a functional fragment thereof
  • a regulatory element in an expression cassette results in selective gene expression in PV cells, or PV cells in the CNS, or PV neurons, wherein about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the cells expressing the transgene are PV positive.
  • an expression cassette or a gene therapy comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more regulatory elements as described in TABLE 1, e.g., SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein are operably linked to any transgene in an expression cassette.
  • the expression cassette is a gene therapy.
  • the expression cassette is part of a vector or a plasmid, e.g., a viral vector or rAAV vector.
  • the expression cassette is part of AAV1, AAV8, AAV9, or AAVDJ or a variant or hybrid thereof.
  • the expression cassette comprises one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein operably linked to a transgene, wherein the transgene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KAV3.2, KV3.3, STXBP1, DNA binding protein (e.g., transcriptional modulator of an endogenous gene), or a variant or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • regulatory elements increase the selective expression of the transgene in PV neurons as compared to non-PV CNS cell-types.
  • such regulatory elements increase the expression of the transgene selectively in a target cell type, such as a PV neuron.
  • the target cell type is a PV cell.
  • Techniques contemplated herein for gene therapy of somatic cells include delivery via a viral vector (e.g., retroviral, adenoviral, AAV, helper-dependent adenoviral systems, hybrid adenoviral systems, herpes simplex, pox virus, lentivirus, and Epstein-Barr virus), and non-viral systems, such as physical systems (naked DNA, DNA bombardment, electroporation, hydrodynamic, ultrasound, and magnetofection), and chemical system (cationic lipids, different cationic polymers, and lipid polymers).
  • a viral vector e.g., retroviral, adenoviral, AAV, helper-dependent adenoviral systems, hybrid adenoviral systems, herpes simplex, pox virus, lentivirus, and Epstein-Barr virus
  • non-viral systems such as physical systems (naked DNA, DNA bombardment, electroporation, hydrodynamic, ultrasound, and magnetofection), and chemical system (cationic lipids, different cati
  • AAV vectors typically have a packaging capacity of ⁇ 4.8 kb
  • lentiviruses typically have a capacity of ⁇ 8 kb
  • adenoviruses typically have a capacity of ⁇ 7.5 kb
  • alphaviruses typically have a capacity of ⁇ 7.5 kb.
  • Some viruses can have larger packaging capacities, for example herpesvirus can have a capacity of >30 kb and vaccinia a capacity of ⁇ 25 kb.
  • Advantages of using AAV for gene therapy include low pathogenicity, very low frequency of integration into the host genome, and the ability to infect dividing and non-dividing cells.
  • the present disclosure contemplates the use of regulatory elements that are shorter than 2.5 kb, 2 kb, 1.5 kb, 1 kb, 900 bp, 800 bp, 700 bp, 600 bp, 500 bp, 400 bp, 300 bp, 200 bp, 150 bp, or 110 bp, but at least 10 bp, 50 bp or 100 bp in length.
  • the size of the combined regulatory element is about 2500 bp, 2000 bp, 1500 bp, 1400 bp, 1300 bp, 1200 bp, 1100 bp, or 1000 bp.
  • each combined regulatory element has a total length of about 100 bp, 200 bp, 300 bp, 400 bp, 500 bp, 600 bp, 700 bp, 800 bp, 900 bp, 1000 bp, 1100 bp, 1200 bp, 1300 bp, 1400 bp, 1500 bp, 1600 bp, 1700 bp, 1800 bp, 1900 bp, 2000 bp, 2100 bp, 2200 bp, 2300 bp, 2400 bp, or 2500 bp.
  • the size of a combined RE has a total length of about 200 bp-3000 bp, 200 bp-2500 bp, 200 bp-2100 bp, 500 bp-2500 bp, 1000 bp-2500 bp, 1500 bp-2500 bp, 1500b-2000 bp, or 2000 bp-2500 bp.
  • a regulatory element of the disclosure is preferably (i) one that selectively drives expression in a cell-type of interest, such as PV cells; (ii) includes a human derived sequence, and (iii) is smaller than 2.5 kb, 2 kb, 1.5 kb, or 1 kb.
  • an expression cassette which can be a circular or linear nucleic acid molecule.
  • an expression cassette is delivered to cells (e.g., a plurality of different cells or cell types including target cells or cell types and/or non-target cell types) in a vector (e.g., an expression vector).
  • a vector can be an integrating or non-integrating vector, referring to the ability of the vector to integrate the expression cassette and/or transgene into a genome of a cell.
  • Either an integrating vector or a non-integrating vector can be used to deliver an expression cassette containing a transgene operably linked to a regulatory element.
  • vectors include, but are not limited to, (a) non-viral vectors such as nucleic acid vectors including linear oligonucleotides and circular plasmids; artificial chromosomes such as human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), and bacterial artificial chromosomes (BACs or PACs); episomal vectors; transposons (e.g., PiggyBac); and (b) viral vectors such as retroviral vectors, lentiviral vectors, adenoviral vectors, and AAV vectors.
  • Viruses have several advantages for delivery of nucleic acids, including high infectivity and/or tropism for certain target cells or tissues.
  • a virus is used to deliver a nucleic acid molecule or expression cassette comprising one or more regulatory elements, as described herein, operably linked to a transgene.
  • viral gene therapy vectors or gene delivery vectors include the ability to be reproducible and stably propagated and purified to high titers; to mediate targeted delivery (e.g., to deliver the transgene specifically to a tissue or organ of interest without widespread vector dissemination elsewhere or off-target delivery); and to mediate gene delivery and/or transgene expression without inducing harmful side effects or off-target effects.
  • targeted expression or tissue/cell type selective expression can be achieved by placing the transgene under the control of a cell-type-selective regulatory element, e.g., one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto, an enhancer, promoter, stability element, UTR, or a combination thereof.
  • viral particles containing a viral vector can be designed to infect many different cell types but expression of the transgene is enhanced and/or optimized in a cell type of interest (e.g.
  • PV neurons vascular endothelial cells
  • expression of the transgene is reduced and/or minimized in other non-target cell types (e.g., non-PV CNS cells).
  • the differential expression of the transgene in different cell types can be controlled, engineered, or manipulated using different transcription factors or regulatory elements that are selective for one or more cell types.
  • one or more regulatory elements such as a promoter or enhancer, or a combination thereof, are operably linked to a transgene to drive tissue- or cell-selective expression of the transgene.
  • one or more regulatory elements used in a gene therapy or a vector drive gene expression in a cell type selective manner i.e., confer selective gene expression in a target cell, cell type, or tissue, and/or do not drive gene expression in one or more (e.g., at least one, two, three, or four) off-target cells or cell types.
  • one or more regulatory elements operably linked to a transgene enhances selective expression of the transgene in a target cell, cell type, or tissue, while the one or more regulatory elements suppress transgene expression in off-target cells, cell type, or tissue, or confers significantly lower, de minimis, or statistically lower gene expression in one or more off-target cells, cell types, or tissue.
  • virus-based vectors can be obtained by deleting all, or some, of the coding regions from the viral genome, and leaving intact those sequences (e.g., inverted terminal repeat sequences) that are necessary for functions such as packaging the vector genome into the virus capsid or the integration of vector nucleic acid (e.g., DNA) into the host chromatin.
  • An expression cassette comprising a transgene for example, can be cloned into a viral backbone such as a modified or engineered viral backbone lacking viral genes, and used in conjunction with additional vectors (e.g., packaging vectors), which can, for example, when co-transfected, produce recombinant viral vector particles.
  • additional vectors e.g., packaging vectors
  • an AAV serotype that can cross the blood brain barrier or infect cells of the CNS is preferred.
  • AAV9 or a variant thereof is used to deliver an expression cassette of this disclosure, comprising one or more PV selective regulatory elements operably linked to a transgene.
  • viral gene therapies can be engineered to have tropism for a cell type or tissue of interest over non-target cell types or tissues.
  • viral gene therapies can be engineered to infect and deliver a payload or a therapeutic agent, e.g., a transcriptional modulator or a transgene, to one or more regions, tissues, or cell types within the CNS (e.g., PV cells), while having minimal effects on off-target tissues or cell types (e.g., non-CNS tissue or cell types, non-PV CNS cells).
  • viral gene therapies can be engineered to deliver a transgene across the blood brain barrier and/or target a specific region or tissue within the CNS (e.g., hippocampus) or a cell type within the CNS, e.g., PV cells.
  • an AAV vector or an AAV viral particle, or virion, used to deliver one or more regulatory elements and a transgene into a cell, cell type, or tissue, in vivo or in vitro is preferably replication-deficient.
  • an AAV virus is engineered or genetically modified so that it can replicate and generate virions only in the presence of helper factors.
  • the expression cassette is designed for delivery by an AAV or a recombinant AAV (rAAV).
  • an expression cassette is delivered using a lentivirus or a lentiviral vector.
  • larger transgenes i.e., genes that exceed the cloning capacity of AAV, are preferably delivered using a lentivirus or a lentiviral vector.
  • the AAV used in the compositions and methods described herein can be of any serotype (e.g., AAV1, AAV2, AAV5, AAV8, AAV9, and AAVDJ), including hybrid or chimeric AAV serotypes.
  • AAV is used to deliver and/or express a transgene operably linked to one or more regulatory elements that are selective for PV neurons as compared to non-PV CNS cells.
  • an AAV with a high tropism for CNS cells and/or crosses the blood brain barrier is used.
  • AAV1, AAV8, AAV9, and/or AAVDJ are used to deliver to deliver an expression cassette described herein.
  • an expression cassette comprises one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein operably linked to a transgene that is known to be insufficiently expressed in vivo, such as in a disease or condition associated with haploinsufficiency in the gene.
  • the transgene is a voltage-gated ion channel (e.g., a sodium ion channel or a potassium ion channel), a neurotransmitter regulator, or a subunit or functional fragment thereof.
  • the transgene is a DNA binding protein, an ion channel, a neurotransmitter regulator, or a subunit of the ion channel or neurotransmitter regulator.
  • the transgene is a DNA binding protein that comprises one or more zinc fingers.
  • the DNA binding protein comprises a domain of Cas9, a Cas family protein, nuclease-inactivated Cas9 (or dCas9), a dCas family protein, or a transcriptional activator like effector (TALE).
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene comprises a gene editing protein, e.g., a Cas protein, Cas9.
  • the transgene is a voltage-gated ion channel or a subunit thereof, such as SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, or KV3.3, or a functional fragment or variant thereof.
  • the transgene is an alpha subunit of a sodium ion channel.
  • the transgene is a beta subunit of a sodium ion channel.
  • the neurotransmitter regulator is STXBP1 or a functional fragment or variant thereof.
  • an expression cassette is delivered as a viral vector, such as AAV.
  • the AAV is AAV1, AAV8, AAV9, AAV-DJ, scAAV1, scAAV8, or scAAV9.
  • a gene therapy comprising an expression cassette of this disclosure is administered to a subject in need thereof (e.g., a human patient, a mammal, a transgenic animal, or an animal model).
  • the subject in need thereof has symptoms of, has been diagnosed with, or is at risk of developing Alzheimer's disease, Dravet syndrome, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability, and/or seizures.
  • the subject in need thereof has an insufficient gene expression or a mutation in any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, and STXBP1.
  • a gene therapy such as rAAV9, is used to deliver an expression cassette comprising one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein operably linked a transgene, wherein the transgene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein, or a functional fragment thereof, or a sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, a DNA binding protein, or a functional fragment thereof.
  • the transgene comprises a sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to any one of SEQ ID NOs: 37-43, or a functional fragment thereof, as provided in TABLE 2 below.
  • the transgene is any one of SEQ ID NOs: 36-43, or a functional fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • one or more regulatory elements disclosed herein are operably linked to any one of SEQ ID NOs: 36-43 in an expression cassette, or a functional fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the one or more PV cell selective regulatory elements comprise sequences of SEQ ID NOs: 1-32, a functional fragment or a combination thereof, or sequences comprising at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity thereto.
  • sequence identity is measured by BLAST.
  • such gene therapy is used to treat epilepsies, neurodegeneration, tauopathy, neuronal hypoexcitability, Dravet syndrome and/or Alzheimer's disease.
  • such gene therapy is used to treat epilepsy and/or seizures associated with Dravet syndrome and/or Alzheimer's disease.
  • treatment using a gene therapy described herein results in reduced seizure frequency and/or duration.
  • treatment using a gene therapy described herein results in increased formation of functional sodium ion channels, functional potassium ion channels, or functional neurotransmitter regulatory in vivo.
  • AAV serotypes 1, 8, and/or 9, or a hybrid thereof can be used with an expression cassettes described herein to target selective expression in PV cells.
  • an expression cassette designed for delivery by an AAV comprises a 5′ ITR, one or more cell-type selective regulatory elements, an optional enhancer, an optional minimal promoter, a transgene, optionally one or more introns, an optional polyA signal, and a 3′ ITR.
  • an expression cassette can contain a 5′ ITR, two cell-type selective REs, a basal promoter, a transgene, one or more post-transcriptional RNA regulatory elements, and a 3′ITR.
  • the expression cassette contains a 5′ AAV ITR, an enhancer (e.g., PV cell selective enhancer or one or more combined regulatory elements), a promoter (e.g., one or more PV cell selective promoters or regulatory elements), a transgene (e.g., SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, or a DNA binding protein), a post-transcriptional regulatory element, and a 3′ AAV ITR.
  • the promoter can be PV cell selective, or a constitutive promoter.
  • the transgene is a reporter gene, e.g., a coding sequence for eGFP, RFP, or a fluorescent marker.
  • the transgene is a DNA binding protein that modulates gene expression.
  • the transgene is a therapeutic transgene, e.g., a coding sequence for SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, STXBP1, ora DNA binding protein, or a functional fragment or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the post-transcriptional regulatory element can be any sequence which influences the expression of a protein from an mRNA or stability of an RNA, for example, an intron, an internal ribosome entry site (IRES), or a woodchuck hepatitis virus post-transcriptional regulatory element.
  • the post-transcriptional regulatory element is a combination of two or more post-transcriptional regulatory elements.
  • the expression cassette can be designed for delivery by an optimized therapeutic retroviral vector, e.g., a lentiviral vector.
  • the retroviral vector can be a lentiviral vector comprising a left (5′) LTR; sequences which aid packaging and/or nuclear import of the virus, at least one cell-type selective regulatory element, optionally a lentiviral reverse response element (RRE); optionally a promoter or active portion thereof; a transgene operably linked to one or more regulatory elements; optionally an insulator; and a right (3′) retroviral LTR.
  • the expression cassette comprises one or more cell-type selective regulatory elements disclosed herein. In some cases, the expression cassette comprises two or more regulatory elements combined. In some examples, the expression cassette comprises two or more regulatory elements that are not combined, for example, a promoter upstream of the transgene and an enhancer or stability element located downstream of the transgene.
  • the expression cassette contains a putative cell-type selective regulatory element that has selective activity in a cell type of interest, for example, a putative PV cell selective regulatory element.
  • the expression cassette containing the putative regulatory element can be packaged in a viral vector and transfected into an animal model to assess the activity of the putative cell-type selective regulatory element.
  • a putative cell type selective regulatory element can be assessed in vitro or ex vivo by delivering a vector containing the putative cell type selective regulatory element into a plurality of cells or cell types that include a target cell or cell type, and then comparing the cell type selective activity of the putative regulatory element to a control regulatory element, such as a constitutive promoter or regulatory element, or a previously known regulatory element.
  • selective expression is used to selectively express a therapeutic moiety or a transgene in a cell-type of interest (or tissue-type of interest), such as PV neurons in the CNS.
  • a vector comprising a cell-type selective regulatory element operably linked to a transgene results in an increased selective expression of the transgene in the cell-type of interest as compared to one or more (e.g., at least two, three, four, or five) other cells, cell types, tissues, or tissue types, or results in a preferred expression of the transgene in the cell-type of interest as compared to one or more cells or cell types, e.g., at least one, two, three, four, or five non-target cell types.
  • Any known technique can be used to deliver the regulatory elements and a transgene, or compositions comprising regulatory elements and a transgene, to cells of interest (or a target cell or cell type) to confer or induce in vitro, in vivo, or ex vivo expression of the transgene in a cell-type selective manner.
  • the expression cassettes containing cell-type selective regulatory elements of this disclosure further comprise one or more transgenes.
  • the transgenes can be protein-coding genes.
  • the expression cassette contains a transgene.
  • the transgene can replace an absent or defective gene, or compensate for deficient expression of a protein inside a cell.
  • the transgene can be involved in a cell signaling pathway.
  • a transgene can encode a wild-type protein, a functional fragment thereof, a variant or mutant protein having enhanced therapeutic properties, e.g., enhanced activity.
  • the transgene can encode a DNA binding protein comprising one or zinc finger or a domain of dCas9, an ion channel, such as a potassium ion channel or a sodium ion channel, or a subunit thereof, a neurotransmitter factor or a neurotransmitter regulator.
  • a transgene can encode an ion channel subunit, a variant, or a mutant thereof.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene comprises a gene editing protein, e.g., a Cas protein, Cas9.
  • the regulatory elements disclosed herein can be located at any position within an expression vector or cassette.
  • the regulatory elements can be positioned upstream of an enhancer, downstream of an enhancer but upstream of a promoter, within the 5′ UTR of a transgene, within an intron in the transgene, in the 3′ UTR of the transgene, or downstream of the transgene.
  • one or more regulatory elements are positioned upstream or downstream of the operably linked transgene.
  • a regulatory element of this disclosure results in selective expression of an operably linked transgene at a level that is at least 0.5, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.5, or 3 IU/ml in a target cell type (e.g., PV cells) as measured by ELISA.
  • a regulatory element's ability to increase transgene expression can be assessed in a mouse wherein the total amount of transgene expression in the whole mouse and/or the total number of cell types or tissue types having transgene expression are measured.
  • the activity or expression can be represented as an activity or expression level per unit dose, or normalized to a dose of expression cassette or vector administered or delivered to a cell, mouse, or a subject.
  • expression or activity of a transgene is normalized to an amount of plasmid or DNA (e.g., ⁇ g/kg per mouse), or viral particles (e.g., normalized to an amount of genome copies/kg per mouse or subject) used to allow comparison across different expression vectors or cassettes with or without a regulatory element.
  • selective expression or activity in PV cells assayed can be normalized to a dose of about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or greater than 10 or 720 ⁇ g of expression vector, cassette, or plasmid per mouse.
  • the expression level or activity can be normalized to 10 10 , 10 11 , 10 12 , 10 13 , 10 14 , or 10 15 gc/kg of viral particles containing an expression vector or cassette as disclosed herein per mouse.
  • an expression cassette comprises one or more regulatory elements (e.g., any one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto) disclosed herein operably linked to a transgene to result in cell-type selective expression, or preferential expression, of the transgene in a target cell type over at least one, at least two, at least three, at least four, at least five, or more than five non-target cell types.
  • regulatory elements e.g., any one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto
  • an expression cassette comprises one or more regulatory elements operably linked to a transgene to result in cell-type selective expression or preferential expression in a target cell subtype over at least one, at least two, at least three, at least four, at least five, or more than five non-target subtypes, or all other known subtypes of the cell.
  • the transgene is any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, and STXBP1, or a functional fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the transgene is a DNA binding protein that modulates an endogenous gene (e.g., an endogenous SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.2, KV3.3, or STXBP1).
  • the transgene is any one of SEQ ID NOs: 36-43, or a functional fragment thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the transgene is a transcriptional modulator.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene is a gene editing protein, such as a Cas family protein, Cas9, a zinc finger nuclease, a zinc finger nuclease, or a transcription activator-like effector nuclease.
  • the transgene is a reporter gene or a fluorescent marker.
  • an expression cassette disclosed herein is in a viral vector.
  • an expression cassette disclosed herein is packaged in an rAAV, such as rAAV9 or rAAVDJ.
  • an expression cassette disclosed herein is delivered into a cell as a gene therapy.
  • a gene therapy disclosed herein is delivered into a subject, preferably a human or a mammal.
  • an expression cassette disclosed herein is used to treat a neurological condition or disease, such as epilepsy, a neurodegenerative disease, tauopathy, neuronal hypoexcitability, Dravet syndrome or Alzheimer's disease.
  • an expression cassette (e.g., gene therapy, viral vector, vector, or plasmid) comprises any one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto, operably linked to a transgene.
  • such combined regulatory elements are linked using a linker of 1-50 nucleotides. In some cases, such combined regulatory elements are not linked.
  • two or more regulatory elements are located upstream and/or downstream of the promoter. In some cases, two or more regulatory elements are located upstream and/or downstream of the transgene.
  • an expression cassette comprises one or more regulatory elements disclosed herein, when operably linked to any transgene (e.g., a reporter transgene or a therapeutic transgene), drives selective expression or preferential expression in at least one target cell type at a level that is statistically significantly higher than the expression driven by CAG, EF1 ⁇ , a constitutive promoter, or a non-selective regulatory element when operably linked to the same transgene, or by the same construct without the regulatory elements.
  • any transgene e.g., a reporter transgene or a therapeutic transgene
  • the regulatory elements drive selective expression in the target cell type at a level that is at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 times the expression level by CAG, EF1 ⁇ , a constitutive promoter, or a non-selective regulatory element when operably linked to the same transgene in the target cell type, or by the same construct without the regulatory elements.
  • such cell-type selective expression is assayed using a co-localization assay as described herein.
  • an expression cassette comprising one or more regulatory elements disclosed herein operably linked to any transgene disclosed herein results in selective expression or preferential expression of the transgene in a target cell type over at least one, at least two, at least three, at least four, at least five, or more than five non-target cell types or non-target subtypes.
  • the target cell type is a PV cell.
  • the non-target cell subtypes are at least one, at least two, at least three, or at least four of the non-PV GABAergic subtypes disclosed herein.
  • an expression cassette comprising a regulatory element disclosed herein is selective for PV cells over all non-PV GABAergic cells or all non-PV CNS cells.
  • cell-type selectivity is measured according to a co-localization assay disclosed herein.
  • cell-type selectivity is measured using a mouse that expresses Cre in the target cell type.
  • GABAergic neurons produce gamma aminobutyric acid (GABA), the main inhibitory neurotransmitter in the CNS.
  • GABA is important for reducing neural excitability throughout the nervous system. GABA acts at inhibitory synapses by binding specific transmembrane receptors and causing the opening of ion channels which negatively change the membrane polarization. This generally results in hyperpolarization of the cell and increases the signal required to trigger an action potential. Defects in GABAergic neurons can result in an imbalance between excitatory and inhibitory signaling, and have been implicated in many neurological diseases, including Dravet syndrome, epilepsy, neurodegeneration, tauopathies and Alzheimer's disease.
  • neurological conditions or diseases implicated include a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); and/or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • Parvalbumin is a calcium-binding protein, which is expressed in about 40% of total GABAergic interneurons in the somatosensory cortex.
  • PV cells are generally considered GABAergic cells.
  • GABAergic cells include distinct subtypes of cells, including cells that express PV, SOM, CR, CCK, NPY, VIP, or a combination thereof.
  • PV neurons are particularly relevant for various neurological diseases or conditions, such as Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathies and/or seizures.
  • a PV neuron-associated neurological condition or disease is a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • the target cell is PV cells in the CNS, or GABAergic cells that express PV.
  • PV-expressing interneurons are also called basket cells, which can be further subdivided by size of the cell body (e.g., large basket cell, small basket cell, and nest basket cell), and dendritic and axonal projection.
  • Physiologically, PV-expressing basket cells are often fast-spiking (FS), characterized by a high-frequency train of action potentials (APs) with little adaptation. It is widely accepted that PV basket neurons innervate the soma and proximal dendrites of excitatory pyramidal neurons. Feedforward inhibition mediated through FS PV-expressing basket neurons can be found in several cortical networks including thalamocortical, translaminar, and interareal circuits.
  • FS PV basket neurons strongly inhibit neighboring excitatory pyramidal neurons. It has been shown that PV basket neurons and pyramidal neurons that share common excitatory inputs tend to be reciprocally connected (feedback inhibition). These connections can serve to regulate the precise time window in which the excitatory neurons can generate spikes in response to excitatory drives. In addition, thalamocortical and intracortical excitatory inputs onto FS PV basket neurons are depressed by high frequency stimulation, which mediates activity-dependent feedforward inhibition. PV-expressing basket cells also innervate other interneurons including other basket cells, and are electrically coupled with each other through gap junctions. It has been proposed that this feature may help to generate and maintain cortical network synchronization and oscillation.
  • one or more regulatory elements disclosed herein result in increased selectivity in gene expression in PV neurons as compared to at least one, at least two, at least three, at least four, or at least five non-PV expressing neurons.
  • non-PV cells include all non-PV GABAergic cells.
  • non-PV GABAergic neurons include, but are not limited to, calretinin (CR), somatostatin (SOM), cholecystokinin (CCK), CR+SOM, CR+neuropeptide Y (NPY), CR+vasointestinal polypeptide (VIP), SOM+NPY, SOM+VIP, VIP+choline acetyltransferase (ChAT), CCK+NPY, CR+SOM+NPY, and CR+SOM+VIP expressing cells.
  • any one of CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element that drives gene expression in a non-cell type selective manner can be used for comparison with PV selective regulatory elements, or any cell type selective regulatory elements, disclosed herein.
  • a regulatory element that results in selective expression in PV cells at a level above the expression of a gene operably linked to CAG or EF1 ⁇ control is indicative of selectivity to PV cells.
  • a regulatory element disclosed herein shows selective gene expression in PV cells that is at least 2%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% higher than the PV expression level from a transgene that is operably linked to CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto), and as measured in a co-localization assay.
  • a constitutive promoter e.g.,
  • a regulatory element disclosed herein shows a selective gene expression in PV cells that is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, or at least 10 fold the expression level under CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto) and as measured in a co-localization assay described herein.
  • a constitutive promoter e.g., SV40, CMV, UBC, PGK, and CBA
  • a non-selective regulatory element e.g., SEQ ID NO: 34, or a functional fragment thereof,
  • one or more regulatory elements described herein selectively drive expression of a transgene in a GABAergic cell, such as a GABAergic cell that expresses parvalbumin over at least one other CNS cell type (e.g., at least two, at least three, at least four, at least five non-PV cells, or two or more, three or more, or four or more non-PV cells and/or non-PV GABAergic neurons).
  • a GABAergic cell such as a GABAergic cell that expresses parvalbumin over at least one other CNS cell type (e.g., at least two, at least three, at least four, at least five non-PV cells, or two or more, three or more, or four or more non-PV cells and/or non-PV GABAergic neurons).
  • a target cell type is a GABAergic neuron that expresses parvalbumin, or PV cells.
  • One way of selectively expressing a transgene within a subpopulation of cells in the brain is to use a viral vector comprising a transgene operably linked to a cell-type selective regulatory element, or a regulatory element that is selective (or has selective activity) in the subpopulation of cells in the brain, e.g., PV cells.
  • a viral vector can be selected to have high infectivity without selectivity for a particular cell type, while the regulatory element confers selectivity.
  • a cell-type selective regulatory element can drive expression of a transgene in PV neurons and not in other neurons.
  • the present disclosure involves the use of regulatory elements (i.e., PV cell selective regulatory elements) that selectively drive expression in PV neurons.
  • regulatory elements i.e., PV cell selective regulatory elements
  • GABAergic cells are inhibitory neurons which produce gamma-aminobutyric acid. GABAergic cells can be identified by the expression of glutamic acid decarboxylase 2 (GAD2). Other markers of GABAergic cells include GAD1, NKX2.1, DLX1, DLXS, SST, PV and VIP.
  • a non-PV CNS cell is an excitatory neuron, a dopaminergic neuron, an astrocyte, a microglia, a motor neuron or a vascular cell.
  • a non-GABAergic neuron is a cell that does not express one or more of GAD2, GAD1, NKX2.1, DLX1, DLXS, SST and VIP.
  • a non-PV neuron is a GABAergic neuron that does not express parvalbumin.
  • other CNS cells refer to CNS cell types that have never expressed any of PV, GAD2, GAD1, NKX2.1, DLX1, DLXS, SST and VIP.
  • a regulatory element disclosed herein is selective for a PV expressing cell over at least one, two, three, four, five, or more than five non-PV CNS cell-types.
  • non-PV cell types include non-PV GABAergic cells.
  • the cell type of interest is a PV cell.
  • REs selective for PV cells are referred to as PV cell selective regulatory elements.
  • the PV cell selective regulatory elements disclosed herein include the sequences of SEQ ID NOs: 1-32, or any combination thereof.
  • one or more PV cell selective regulatory elements or one or more regulatory elements disclosed herein are used to increase expression of a transgene in PV-expressing cells by at least 2, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more fold as compared to expression without the regulatory element.
  • a RE in an expression cassette increases gene expression by at least 1.5%, 2%, 5%, 10%, 15%, 20%, or 50%, or more than 1.5%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% as compared to expression without the regulatory elements.
  • compositions and methods of use thereof comprise expression cassettes containing one or more regulatory elements that result in a 10-500% increase in transgene expression, e.g., expression of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KCNC1 (also known as KV3.1), KCNC3 (also known as KV3.3), STXBP1, a DNA binding protein, or a variant or functional fragment thereof, or a protein thereof, as compared to the level without the regulatory elements or as compared to a non-selective regulatory element (e.g., CAG, EF1 ⁇ , a constitutive promoter, or SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto).
  • a non-selective regulatory element e.g., CAG, EF1 ⁇ , a constitutive promoter, or SEQ ID NO: 34, or a
  • the increase in gene expression and/or protein level of any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KCNC1, KCNC3, and STXBP1 is 1.5-5%, 5%-10%, 10-15%, 15-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90%, 90-100%, 100-150%, 150-200%, 250-300%, 300-350%, 350-400%, 400-450%, 450-500%, or 1.5-20%, 20%-50%, 50%-100%, 100-200%, 200-300%, 300-400%, or 400-500% as compared to the level without the expression cassette or regulatory elements.
  • such gene or protein expression is selective in PV cells as compared to an expression cassette comprising a control (e.g., CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element) or a non-cell type selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto).
  • a control e.g., CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), or a non-selective regulatory element) or a non-cell type selective regulatory element (e.g., SEQ ID NO: 34, or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least
  • selectivity of expression in PV cells can be calculated by dividing the number of cells that express both PV and eGFP (the transgene operably linked to one or more regulatory elements) by the total number of cells that express eGFP, and multiplying by 100 to convert into a percentage.
  • PV cell selective regulatory elements as described herein can be highly selective for expression in PV cells.
  • PV cell selective regulatory elements or one or more regulatory elements disclosed herein can exhibit about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than about 99% selectivity for PV neurons.
  • a PV cell selective regulatory element or any regulatory element disclosed herein confers selectivity in expressing a transgene in PV neurons at a level that is statistically higher than a control regulatory element, e.g., EF1 ⁇ or a previously known regulatory element.
  • a control regulatory element e.g., EF1 ⁇ or a previously known regulatory element.
  • the statistical difference between a PV cell selective regulatory element and a control regulatory element is at least 2 fold, 5 fold, 10 fold, 20 fold, or more than 2 fold difference, or more than 5 fold, 10 fold, or 20 fold difference as determined by any one of the methods described herein, such as a co-localization assay.
  • the present disclosure includes regulatory elements that are selective for PV cells.
  • These PV cell selective REs or any cell type selective REs are preferably short, preferably less than about 1100 base pairs, 1000 bp, 900 bp, 800 bp, 700 bp, 600 bp, 500 bp, 400 bp, 300 bp, 200 bp, or less than about 110 bp.
  • the PV cell selective REs or any cell type selective REs can be between 1050 bp and 100 bp, between 100 bp and 500 bp, or between 500 bp and 1050 bp.
  • Some examples of PV cell selective regulatory elements are provided by SEQ ID NOs 1-32, or a functional fragment or combination thereof.
  • PV cell selective regulatory elements contemplated by the present disclosure include sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity to any of these sequences described herein, or a part or fragment of one of the sequences described herein.
  • a PV cell selective regulatory element or any regulatory element disclosed herein has at least about 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more than 99% identity to a sequence described herein, or a fragment of a sequence described herein. In some cases, a PV cell selective regulatory element has at least about 80% identity to at least about 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% of a sequence described herein, or a functional fragment thereof.
  • a PV cell selective regulatory element comprises at least 80% identity to any one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a PV cell selective regulatory element has 90% identity to 50% or more of a sequence of SEQ ID NOs: 1-22.
  • a PV-selective regulatory element is a functional fragment of any of SEQ ID NOs: 1-32 or a combination thereof.
  • the functional fragment is able to selectively express a transgene in PV cells with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or greater than 95% selectivity of expression in PV cells.
  • two or more PV cell selective regulatory elements of this disclosure or any two or more regulatory elements disclosed herein are combined to form combination regulatory elements.
  • two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more PV cell selective regulatory elements, or a plurality of regulatory elements disclosed herein are combined.
  • SEQ ID NO: 31 is a combination of SEQ ID NOs 1 and 23-29.
  • SEQ ID NO: 32 is a combination of SEQ ID NO: 8 and SEQ ID NO: 23-29.
  • fragments of two or more PV-selective regulatory elements can be combined to form a combination regulatory element.
  • 50% of SEQ ID NO: 1 can be combined with 30% of SEQ ID NO: 8 and 90% of SEQ ID NO: 30 to form a combination regulatory element.
  • one or more PV cell selective regulatory elements of this disclosure or any one or more regulatory elements disclosed herein selectively express an operably linked transgene in PV neurons as compared to one or more other CNS cell types.
  • This selective expression can be quantified by counting the number of PV neurons which express detectable levels of the linked transgene as a percentage of the total number of cells expression the transgene, including the number of non-PV neurons which express the transgene.
  • selectivity of a PV regulatory element in a particular cell type or target cell can be determined by measuring and/or comparing the number of PV neurons (or target cells) expressing the transgene that is operably linked to the regulatory element relative to the number of non-target cell types that express the transgene (or the total number of cells expressing the transgene).
  • PV cell selective regulatory elements can exhibit about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than about 99% selectivity for PV neurons, PV neurons in the CNS, or GABAergic neurons that also express PV.
  • one or more regulatory elements of this disclosure can exhibit about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than about 99% selectivity for PV neurons as compared to CAG or EF1 ⁇ or non-cell-type selective element, or as compared to non-PV CNS cells, or as compared to at least one, at least two, at least three, at least four, or at least five other non-PV GABAergic neuronal sub-types in the CNS.
  • a PV selective regulatory element confers selectivity in expressing a transgene in PV neurons at a level that is statistically higher than a control regulatory element, e.g., CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), a non-selective regulatory element, or a previously known regulatory element.
  • a control regulatory element e.g., CAG, EF1 ⁇ , a constitutive promoter (e.g., SV40, CMV, UBC, PGK, and CBA), a non-selective regulatory element, or a previously known regulatory element.
  • the statistical difference between a PV cell selective RE and a control element is at least 2 fold, 5 fold, 10 fold, 20 fold, or more than 2 fold difference, or more than 5 fold, 10 fold, or 20 fold difference as determined by any one of the methods described herein.
  • the selectivity in PV is measured using a co-localization assay as described
  • the cell-type selective regulatory elements described herein are useful for selectively modulating expression of a transgene in a CNS cell type compared to other CNS cell types.
  • the cell-type selective regulatory elements described herein can be useful for selectively modulating expression of a transgene in PV cells over other CNS cells, including other types of neurons.
  • selective expression of a transgene in a target cell type and/or minimized expression of the transgene in a non-target cell type can be desired. Expression of the transgene in an unintended cell-type (e.g., non-target cell type) can result in an adverse effect to the subject.
  • transgene in an unintended cell-type can counteract the therapeutic effect of the transgene in the intended cell type.
  • a transgene intended for expression in PV cells can have a negative effect for the subject if expressed in glutamatergic neurons.
  • the cell-type selective regulatory elements described herein can be used in expression cassettes to ensure appropriate expression of a transgene and/or to reduce off-target effects of gene therapy.
  • the cell-type selective regulatory elements herein can be used in gene expression cassettes whereby they are operably linked to one or more transgenes. Such gene expression cassettes are used to deliver transgenes into cells for expression.
  • the expression cassette can contain a cell-type selective regulatory element as described herein, a combination of cell-type selective regulatory elements, or a fragment of a cell-type selective regulatory element as described herein operably linked to a transgene.
  • the expression cassettes herein include one or more cell-type selective regulatory elements operably linked to a transgene, whereby the two do not function together in their endogenous context in vivo.
  • a transgene for sodium ion channel beta subunit such as SCN1B
  • SCN1B can be operably linked to one or more regulatory elements that do not function in the same context in vivo, or do not detectably drive expression of SCN1B endogenously.
  • a nucleic acid cassette can include a neurotransmitter regulator, such as STXBP1, operably linked to a regulatory element that does not function in the same context endogenously or in vivo, or is not in the same open reading frame, or is not on the same human chromosome, or does not detectably drive expression of STXBP1 in vivo.
  • a cell-type selective regulatory element is linked to a transgene, wherein the cell-type selective regulatory element does not regulate the endogenous gene corresponding to the transgene in vivo.
  • cell-type selective regulatory elements disclosed herein are derived from sequences isolated from a human chromosomal locus different from a locus of a native gene corresponding to the transgene.
  • an expression cassette comprises cell-type selective regulatory element(s) having a sequence derived from a chromosome different from the chromosome corresponding to the transgene on the same cassette.
  • regulatory element(s) of the disclosure and a transgene that are operably linked in an expression cassette are derived from sequences located more than 20 kb apart in the human genome, or at distal genomic locations.
  • the two or more regulatory elements can have sequences located more than 5 kb apart, more than 10 kb apart, more than 15 kb apart, or more than 20 kb apart in the human genome, or wherein the two or more regulatory elements do not interact with each other naturally in the genome.
  • an expression cassette comprising PV-selective regulatory elements can exclude known sequences derived from hSyn1 or GAD2 promoter sequences.
  • the PV-selective regulatory elements do not comprise the full promoter sequence of any one of the GAD2, GAD1, SYN1, NKX2.1, DLX1, DLXS, SST and VIP promoters.
  • the PV-selective regulatory elements do not comprise more than 500 contiguous base pairs of sequence derived from the promoter or one or more of GAD2, GAD1, SYN1, NKX2.1, DLX1, DLX5, SST and VIP.
  • the PV-selective regulatory elements do not comprise sequences which are within 1 kb, 2 kb, 3 kb, 4 kb, 5 kb, 6 kb, 7 kb, 8 kb, 9 kb, or 10 kb of the transcription start site of any one of GAD2, SYN1, NKX2.1, DLX1, DLX5, SST, and VIP.
  • a transgene is useful to treat a disease associated with a specific cell type of interest.
  • a cell type of interest is a neuron, an inhibitory neuron, a GABAergic neuron, or a PV neuron.
  • a transgene is any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, and STXBP1.
  • a transgene is a DNA binding protein that modulates expression of a gene (e.g., a transcriptional activator or a transcriptional repressor that modulates expression of an endogenous gene).
  • a transgene is a gene editing protein, such as a zinc finger nuclease, a transcription activator-like effector nuclease, a Cas family protein.
  • a transgene is a reporter gene or a detectable marker, such as eGFP, tdTomato, or RFP.
  • a transgene is a Cas protein, such as Cas9.
  • Transgenes useful to treat a condition associated with PV neuron cells can be incorporated in a vector, nucleic acid cassette, or method as described herein.
  • Transgenes used herein generally do not contain introns, or do not contain more than one intron.
  • a transgene can be obtained from a cDNA sequence rather than from genomic sequence.
  • transgenes can contain some, or all, of their endogenous introns. In some examples, such a transgene encodes for a DNA binding domain or an ion channel.
  • DNA binding domains that can be encoded for in the expression cassettes of this disclosure include zinc fingers, Cas9, a Cas family protein, dCas9, a dCas family protein or a transcriptional activator like effector (TALE).
  • TALE transcriptional activator like effector
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene comprises a gene editing protein, e.g., a Cas protein, Cas9.
  • the transgene is a subunit or a component of an ion channel or a membrane protein, or a gene associated with a neurological condition or disease disclosed herein.
  • ion channel transgenes which can be used in the expression cassettes of this disclosure include voltage gated and ligand gated ion channels. Voltage gated ion channels include sodium channels, calcium channels, potassium channels, and proton channels. In some instances, the transgene codes for a subunit of a voltage gated sodium channel. Examples of voltage gated sodium channel subunits include SCN1B (NM_001037.4), SCN1A (NM_001165963.1), and SCN2B, (NM_004588.4).
  • the transgene codes for a subunit of a voltage gated potassium channel.
  • voltage gated sodium channel subunits include KCNC1 (NM_001112741.1), and KCNC3 (NM_004977.2).
  • a transgene is any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, a variant and a functional fragment thereof.
  • a transgene is a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% sequence identity to any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, a variant or a functional fragment thereof. In some cases, such sequence identity is measured using BLAST.
  • an expression cassette disclosed herein comprises one or more PV-selective regulatory elements or one or more regulatory elements of this disclosure operably linked to a transgene having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% sequence identity to a sequence encoding any one of SEQ ID NOs: 37-43, or a functional fragment or variant thereof, or the GenBank sequences corresponding to SEQ ID NOs: 37-43.
  • an expression cassette disclosed herein comprises a transgene having a sequence according to (i) a sequence of SEQ ID NOs: 37-43, or (ii) a functional fragment thereof, or (iii) a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% sequence identity to (i) or (ii).
  • the transgene is a neurotransmitter regulator, or a variant or functional fragment thereof.
  • a neurotransmitter regulator may be involved in regulating production or release of a neurotransmitter in the CNS.
  • a neurotransmitter regulator may assist with synaptic fusion to release neurotransmitters.
  • An example of a neurotransmitter regulator is STXBP1 (NM_001032221.3) or a functional fragment thereof, or a sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the transgene may also be a subunit of a neurotransmitter regulator.
  • an expression cassette of this disclosure can contain an AAV2 5′ ITR, a PV-selective enhancer, a PV-selective promoter, or a combination of one or more PV-selective promoters and enhancers, a cDNA of SCN1B, a WPRE, a hGH polyA signal, PV-selective regulatory element, and an AAV2 3′ITR.
  • the expression cassette comprises an AmpR promoter, and AmpR coding sequence, a bacterial origin of replication, an AAV2 ITR, SEQ ID NO: 8, SEQ ID NOs: 23-29, a transgene (coding sequence, a WPRE, a human growth hormone poly A signal, a AAV2 ITR, and an fl origin.
  • an expression cassette of this disclosure can contain an AAV2 5′ ITR, an enhancer, a promoter, a transcriptional activator of the endogenous SCN1A gene, a WPRE, a hGH polyA signal, a regulatory element, and an AAV2 3′ITR.
  • an expression cassette comprises AAV2 5′ ITR, a promoter, an intronic element, transcriptional modifier, synthetic polyA, and an AAV2 3′ ITR.
  • an expression cassette of this disclosure can contain an AAV2 5′ ITR, a PV-selective enhancer, a PV-selective promoter, a sequence encoding a transcriptional activator of SCN1A or SCN1B, a WPRE, a hGH polyA signal, a PV-selective regulatory element or any of the regulatory elements disclosed herein, and an AAV2 3′ITR.
  • An expression cassette comprising one or more regulatory element of this disclosure can be used to treat a medical condition.
  • an expression cassette containing a regulatory element of this disclosure is used to treat a neurological condition or a neurodegenerative condition.
  • the neurological condition can be caused by a known genetic event or may have an unknown cause.
  • the neurological condition can be a disease associated with PV neurons.
  • the neurological condition can be a disease associated with inhibitory neurons, such as PV neurons.
  • Diseases or conditions associated with PV neurons can be treated by delivering an expression cassette carrying a transgene and one or more PV cell-selective regulatory elements or any of the regulatory elements as described herein to a cell in vivo.
  • expression cassettes comprising a transgene operably linked to one or more PV cell-selective regulatory elements or any of the regulatory elements disclosed herein can be used to treat Dravet syndrome, Alzheimer's disease, epilepsy, a neurodegenerative disorder, tauopathy, neuronal hypoexcitability and/or seizures.
  • an expression cassette of this disclosure is used to treat a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., Dravet syndrome, chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); and/or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • Dravet syndrome cases Majority of Dravet syndrome cases is associated with mutations in the SCN1A and/or SCN2A genes. Mutations or abnormalities in SCN1A has also been associated with seizure disorders, epilepsy, autism, familial hemiplegic migraine type 3 (FHM3), genetic epilepsy with febrile seizures plus (GEFS+), and effectiveness of certain anti-seizure medications. For instance, ICS5N+5G>A mutation in SCN1A is associated with the maximum safe amount (dose) of the anti-seizure drugs phenytoin and carbamazepine.
  • amyloid ⁇ involving many peptides and proteases that can affect excitability of neurons, causing seizures and downregulation of the Nav1.1 sodium channel in PV neurons.
  • Diseases associated with dysfunctional PV neurons such as those due to loss of function mutations in SCN1A or Nav1.1 include: Dravet syndrome, Ohtahara syndrome, epilepsy, early infantile epileptic encephalopathy 6 (EIEE6), familial febrile seizures 3A (FEB3A), intractable childhood epilepsy with generalized tonic-clonic seizures (ICEGTC), migraine, familial hemiplegic 3 (FHM3), Panayiotopoulos syndrome, familial atrial fibrillation 13 (ATFB13), generalized epilepsy with febrile seizures plus type 1 (gefs+ type 1), Brugada syndrome, nonspecific cardiac conduction defect, generalized epilepsy with febrile seizures plus, benign familial infantile seizures, early infantile epileptic encephalopathyll (EIEE11), benign familial infantile epilepsy, neurodegeneration, tauopathies and Alzheimer's disease.
  • Dravet syndrome Dravet syndrome, Ohtahara syndrome, epilepsy, early infantile epileptic
  • the neurological condition is Dravet syndrome.
  • Dravet syndrome is associated with mutations in the SCN1A and/or SCN2A genes.
  • one or more regulatory elements of this disclosure are used in a gene therapy or an expression cassette to treat a neurological condition or disease associated with PV neurons, e.g., a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., Dravet syndrome, chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • a psychiatric disorder e.g., schizophrenia, obsessive compulsive disorder, addiction, depression
  • one or more regulatory elements of this disclosure are used to treat Dravet syndrome and/or Alzheimer's disease (e.g., in an expression cassette, a vector, or a gene therapy).
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • Methods and compositions of this disclosure can be used to treat a subject who has been diagnosed with a disease, for example, a neurological or neurodegenerative disease.
  • the subject can be a patient suffering from a form of epilepsy.
  • the subject is a patient with Dravet syndrome.
  • the subject can be a patient suffering from a neurodegenerative disease, for example, a patient with Alzheimer's disease.
  • epilepsy, encephalopathy, and/or seizures are associated with a genetic mutation in SCN8A.
  • a genetic mutation in SCN8A can give rise to epilepsy syndromes, e.g., Dravet syndrome.
  • a genetic mutation in STXBP1 is associated with encephalopathy with epilepsy, characterized by recurrent seizures.
  • a subject treated with one or more compositions described herein is one diagnosed with a mutation or genetic aberration in an ion channel or a neurotransmitter regulator (e.g., a syntaxin binding protein).
  • a neurotransmitter regulator e.g., a syntaxin binding protein
  • mutations include mutations in SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KCNC1, KCNC3, and/or STXBP1, or combination thereof.
  • the expression cassette containing a cell-type selective regulatory element as described herein can be delivered to a subject to treat or prevent a disease with symptoms associated with a specific cell type.
  • an expression cassette comprising a transgene operably linked to one or more PV cell selective regulatory element is delivered to a subject who has symptoms, or is at risk of developing symptoms, associated with PV neurons.
  • the treatment can be administered to a subject with, or at risk of developing, Dravet syndrome.
  • Symptoms associated with Dravet syndrome include seizures, memory defects, developmental delay, poor muscle tone and/or cognitive problems.
  • Treatment with an expression cassette of this disclosure can result in an improvement of one or more symptoms, such as a reduction in number, duration, and/or intensity of seizures.
  • Administration of a gene therapy as described herein to a subject at risk of developing Dravet syndrome can prevent the development of or slow the progression of one or more symptoms.
  • the treatment may be administered to a subject suffering from Alzheimer's disease.
  • Symptoms associated with Alzheimer's disease include short term memory loss, cognitive difficulties, seizures, and difficulties with language, executive functions, perception (agnosia), and execution of movements (apraxia).
  • Treatment with an expression cassette of this disclosure can result in an improvement of one or more Alzheimer's disease symptoms, such as a reduction in progression of memory loss, or the prevention of one or more symptoms.
  • the treatment can result in a correction of high gamma power brain activity.
  • the treatment can result in a decrease in seizure frequency and/or seizure severity, or a decrease in high gamma power activity by 10%, 20%, 30%, 40%, 50%, 60%, or 70%.
  • the treatment can result in an improvement in cognitive function. Learning and/or memory can be improved by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or more than 100%.
  • Methods and compositions of this disclosure can be used to treat a subject who is at risk of developing a disease.
  • the subject can be known to be predisposed to a disease, for example, a neurological disease or a disease associated with epilepsy, seizures, and/or encephalopathy.
  • the subject can be predisposed to a disease due to a genetic event, or due to known risk factors.
  • a subject can carry a mutation in SCN1A which is associated with Dravet syndrome.
  • the subject can be predisposed to a disease such as Alzheimer's disease due to the age of the subject.
  • the treatment can result in a decrease or cessation of symptoms.
  • treatment can improve learning, memory, cognitive function, and/or motor function; reduce frequency and/or duration of seizures; and/or reduce temperature sensitivity (or increase the temperature threshold for triggering a seizure).
  • the target cell type of a gene therapy or expression cassette disclosed herein is a PV cell.
  • the non-target cell subtypes are at least one, at least two, at least three, or at least four of the non-PV GABAergic subtypes disclosed herein.
  • an expression cassette comprising a regulatory element disclosed herein is selective for PV cells over all non-PV CNS cells.
  • cell-type selectivity is measured according to a co-localization assay disclosed herein.
  • cell-type selectivity is measured using a mouse that expresses Cre in the target cell type.
  • the treatment does not result in an adverse reaction for the subject.
  • Treatment with a gene therapy containing a PV-selective regulatory element can cause fewer, or less severe, adverse reactions in a subject than treatment with a similar gene therapy containing the same transgene linked to a non-selective regulatory element.
  • any expression cassette disclosed herein can be adapted for or used in a gene therapy (e.g., rAAV or rAAV9 gene therapy) to treat any one or more of Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizure.
  • a gene therapy e.g., rAAV or rAAV9 gene therapy
  • a gene therapy comprises any one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto, operably linked to a transgene.
  • a gene therapy comprises an expression cassette of this disclosure.
  • a gene therapy comprises one or more regulatory elements disclosed herein operably linked to any transgene (e.g., a reporter transgene or a therapeutic transgene) such that the regulatory elements drive selective expression or preferential expression in at least one target cell type at a level that is statistically significantly higher than the expression driven by CAG or EF 1a or a non-selective regulatory element when operably linked to the same transgene, or by the same construct without the regulatory elements.
  • any transgene e.g., a reporter transgene or a therapeutic transgene
  • the regulatory elements drive selective expression in the target cell type at a level that is at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 times the expression level by the CAG, EF1 ⁇ , a constitutive promoter, or a non-selective regulatory element when operably linked to the same transgene in the target cell type, or by the same construct without the regulatory elements.
  • the transgene is any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, and a functional fragment thereof.
  • the transgene is a DNA binding protein that modulates expression of an endogenous gene, such as a transcriptional modulator, a transcriptional activator, or a transcriptional repressor.
  • the transgene is a DNA binding protein that comprises a DNA binding domain of a DNA binding protein or a DNA cleaving protein (e.g., a nuclease, a restriction enzyme, a recombinase, etc.) wherein the DNA cleaving domain or nuclease domain has been deactivated, e.g., a nuclease-deactivated Cas (dCas), a deactivated transcription activator-like effector nuclease, or a nuclease-deactivated zinc finger protein.
  • the DNA binding domain is linked to a transcriptional modulating domain (e.g., a transcriptional activator or repressor domain).
  • the transgene is a gene editing protein, such as a zinc finger nuclease or a transcription activator-like effector nuclease.
  • a transgene is a reporter gene or a detectable marker, such as eGFP, tdTomato, or RFP.
  • a transgene is a Cas protein, such as Cas9.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat a neurological condition or disease.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat a neurological condition or disease, wherein the expression cassette comprises any one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto, operably linked to a transgene.
  • the transgene is any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, a DNA binding protein and a functional fragment thereof.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat Dravet syndrome.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat Alzheimer's disease.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat epilepsy and/or seizure symptoms associated with Dravet syndrome and/or Alzheimer's disease.
  • treating any one of Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizures comprises delivering or administering a gene therapy of this disclosure to a cell of a subject in need thereof.
  • the subject in need thereof is at risk for or has any one of Dravet syndrome, Alzheimer's disease, epilepsy, and/or seizures.
  • the subject is a child or a minor.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat an infant, a child, or a minor diagnosed with or is at risk of developing Dravet syndrome.
  • a gene therapy comprising an expression cassette disclosed herein is used to treat a subject comprising a mutation or a genetic defect in any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, and STXBP1.
  • the present disclosure provides a method of treating any one of Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizures, comprising administering a gene therapy into a cell of a subject, wherein the gene therapy comprises an expression cassette disclosed herein.
  • such expression cassette comprises any one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto, operably linked to a transgene, wherein the transgene is any one of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, and a functional fragment thereof.
  • the transgene is a subunit of a sodium ion channel or a potassium ion channel.
  • the transgene is a syntaxin binding protein.
  • the transgene is a transcriptional modulator, e.g., a transcriptional activator or a transcriptional repressor. In some cases, the transgene is a transcriptional modulator that modulates the expression of an endogenous gene (e.g., SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or STXBP1). In some cases, a transgene is a gene editing protein, such as a zinc finger nuclease or a transcription activator-like effector nuclease. In some cases, the transgene is a Cas protein, such as Cas9.
  • a transcriptional modulator e.g., a transcriptional activator or a transcriptional repressor.
  • the transgene is a transcriptional modulator that modulates the expression of an endogenous gene (e.g., SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV
  • the present disclosure provides a method for modifying any gene therapy designed for treating Dravet syndrome, Alzheimer's disease, epilepsy, neurodegeneration, tauopathy, neuronal hypoexcitability and/or seizures by adding one or more regulatory elements disclosed herein to improve the cell-type selectivity of the gene therapy.
  • the gene therapy is an rAAV gene therapy.
  • treatment with an expression cassette disclosed herein reduces seizure duration and/or frequency, e.g., seizures associated with Dravet syndrome, by at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% as compared to an untreated control or as compared to the level before treatment.
  • seizure duration and/or frequency e.g., seizures associated with Dravet syndrome
  • treatment with an expression cassette disclosed herein reduces high gamma power activity (e.g., high gamma power activity associated with Alzheimer's disease) by at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% as compared to an untreated control or as compared to the level before treatment.
  • high gamma power activity e.g., high gamma power activity associated with Alzheimer's disease
  • the present disclosure provides a nucleic acid cassette of this disclosure comprises one or more regulatory elements operably linked to a transgene that result in selective expression in a target cell type over one or more non-target cell types, e.g., selective expression in PV neurons in the CNS over one or more non-PV CNS cell types.
  • each of the regulatory elements comprises (i) a sequence of SEQ ID NOs: 1-32, (ii) a functional fragment or a combination thereof, or (iii) a sequence with at least 80% sequence identity to (i) or (ii).
  • the percent sequence identity can be measured using BLAST.
  • at least one of the regulatory elements is human derived.
  • At least one of the regulatory elements is derived from a non-human mammal. In some cases, the regulatory elements are non-naturally occurring. In some cases, the regulatory elements result in a selectivity of expression in PV cells that is greater than expression of the transgene operably linked to CAG or EF la or a non-selective regulatory element as measured by a co-localization assay.
  • a transgene is any one or more of SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, and STXBP1, or a functional fragment hereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • a transgene is a DNA binding protein that modulates expression of a gene (e.g., an endogenous gene such as SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, and STXBP1), such as transcriptional modulator, transcriptional activator, or transcriptional repressor.
  • a transgene is a gene editing protein, such as a zinc finger nuclease or a transcription activator-like effector nuclease.
  • a transgene is a reporter gene or a detectable marker, such as eGFP, tdTomato, or RFP.
  • a transgene is a Cas protein, such as Cas9.
  • the regulatory elements result in selective expression in PV cells at a level that is at least 0.5 fold, at least 0.6 fold, at least 0.7 fold , at least 0.8 fold , at least 0.9 fold , at least 1.1 fold, at least 1.2 fold, at least 1.3 fold, at least 1.4 fold, at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 6 fold, at least 7 fold, at least 8 fold, at least 9 fold, at least 10 fold, at least 11 fold, at least 12 fold, at least 13 fold, at least 14 fold, at least 15 fold, at least 16 fold, at least 17 fold, at least 18 fold, at least 19 fold, at least 20 fold, at least 25 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 60 fold, at least 70 fold, at least 80 fold, at least 90 fold, at least 100 fold as compared to that of a CAG or EF 1 a or a non-selective regulatory element, as measured by the co-
  • a fold difference refers to the fold difference between the percentage of eGFP+, PV+ cells that result from one or more regulatory elements and that of a non-selective regulatory element.
  • the co-localization assay is an immunohistochemical assay, as described below in Example 5. In some instances, a co-localization assay is performed using a commercially available anti-PV antibody.
  • the transgene encodes an ion channel subunit, a neurotransmitter regulator, or a variant or a functional fragment thereof.
  • the ion channel subunit is an alpha subunit or a beta subunit of a sodium ion channel or a subunit of a potassium ion channel.
  • the transgene is any one of (i) SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or a DNA binding protein; (ii) a functional fragment thereof; or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the neurotransmitter regulator is (i) STXBP1, (ii) a functional fragment thereof, or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the regulatory elements and the operably linked transgene are located on different chromosomes.
  • the regulatory elements combined are less than 2.5 kb, less than 1.5 kb, less than 1 kb, or less than 500 bp in size.
  • the non-PV cells comprise any one or more of non-PV CNS cell types, including but not limited to excitatory neurons, dopaminergic neurons, astrocytes, microglia, or motor neurons.
  • the nucleic acid cassette is a linear construct.
  • the nucleic acid cassette is a vector.
  • the nucleic acid cassette is a plasmid.
  • the vector is a viral vector.
  • the viral vector is an adeno-associated virus (AAV) vector.
  • the AAV vector is AAV1, AAV8, AAV9, scAAV1, scAAV8, or scAAV9.
  • the viral vector is a lentiviral vector.
  • the regulatory elements contain less than 600 bp of contiguous sequence from within 10 kb of the transcription start site of GAD2, GAD1, SYN1, NKX2.1, DLX1, DLXS/6, SST, PV, and/or VIP.
  • a regulatory element is less than 2050 bp, 2000 bp, 1900 bp, 1800 bp, 1700 bp, 1600 bp, 1500 bp, 1400 bp, 1300 bp, 1200 bp, 1100 bp, 1000 bp, 900 bp, 800 bp, 700 bp, 600 bp, 500 bp, 400 bp, 300 bp, 200 bp, 100 bp, 90 bp, 80 bp, 70 bp, 60 bp, 50 bp, 40 bp, 30 bp, 20 bp, 10 bp, or 5 bp.
  • an expression cassette comprises a transgene that is larger than a typical transgene size in a conventional viral vector, e.g., AAV.
  • an expression cassette of any embodiment disclosed herein comprises a transgene that is at least 1 kb, 1.5 kb, 2 kb, 2.5 kb, 3 kb, 3.5 kb, 4 kb, 4.5 kb, 5 kb, 5.5 kb, 6 kb, 6.5 kb, 7 kb, 7.5 kb, or 8 kb.
  • any embodiment disclosed herein comprises an expression cassette (e.g., AAV) that comprises a transgene that is more than 1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, or 4 kb in size.
  • any embodiment disclosed herein can further comprise one or more heterologous nucleic acid sequence or element.
  • a method of treating a neurological disorder or condition in a subject in need thereof comprises delivering a therapeutically effective amount of a nucleic acid cassette described herein.
  • a method of increasing selective expression of a transgene in PV neurons comprises contacting a cell with a nucleic acid cassette described herein.
  • a method of any embodiment disclosed herein is used to treat a neurological condition or disease, e.g., a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., Dravet syndrome, chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • a psychiatric disorder e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis
  • an autism spectrum disorder e.g., Fragile X syndrome, Rett syndrome
  • epilepsy e.g., Dravet syndrome, chronic traumatic ence
  • a method of any embodiment disclosed herein can be used to treat Dravet syndrome.
  • a method of any embodiment disclosed herein can be used to treat Alzheimer's disease.
  • methods and/or compositions of this disclosure can be used to treat any neurological condition or disease associated with seizure and/or epilepsy, and/or wherein PV neurons are implicated.
  • a neurological condition described herein is treated with a gene therapy, preferably one that results in preferential expression in one tissue type or cell type over another, e.g., a PV neuron as determined via a co-localization assay.
  • the gene therapy is an AAV.
  • a method of targeting expression of any transgene to PV neurons in the CNS comprises operably linking one or more of PV neuron selective regulatory elements to a transgene.
  • the regulatory elements comprise one or more sequences of SEQ ID NOs: 1-32, or sequences with at least 80% sequence identity to SEQ ID NOs: 1-32, or a functional fragment thereof.
  • the regulatory elements result in selective expression in PV neurons at a level that is at least 2 fold, at least 5 fold, or at least 7 fold, or at least 10 fold as compared to CAG or EF1 ⁇ or a non-selective regulatory element operably linked to the transgene, as measured by a co-localization assay.
  • the transgene is SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, STXBP1, a DNA binding protein, or a functional fragment thereof.
  • the regulatory elements and the transgene are in an AAV.
  • the AAV is AAV9.
  • a method of treating a neurological condition or disorder in a subject in need thereof comprises contacting a cell with a nucleic acid cassette comprising one or more regulatory elements operably linked to a transgene that results in selective expression in PV neurons over one or more non-PV CNS cells.
  • the regulatory elements comprise one or more of SEQ ID NOs: 1-32, or a functional fragment or a combination thereof, or sequences having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.
  • the transgene is a voltage-gated ion channel subunit, or a variant or a functional fragment thereof.
  • the subunit is a beta subunit of a sodium ion channel.
  • the subunit is an alpha subunit of a sodium ion channel. In some cases, the subunit is of a potassium ion channel.
  • the transgene is any one of (i) SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, a DNA binding protein or STXBP1; (ii) a functional fragment thereof; or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the neurological condition or disorder is associated with a haploinsufficiency or a mutation in any of SCN1A, SCN1B, SCN2B, KV3.1, and KV3.3.
  • the neurological condition or disorder is Dravet syndrome. In some cases, the neurological condition or disorder is Alzheimer's disease. In some cases, the neurological condition or disease is a psychiatric disorder (e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis); an autism spectrum disorder (e.g., Fragile X syndrome, Rett syndrome); epilepsy (e.g., chronic traumatic encephalopathy, generalized epilepsy with febrile seizures plus (GEFS+), epileptic encephalopathy, temporal lobe epilepsy, focal epilepsy, tuberous sclerosis); or neurodegeneration (e.g., Alzheimer's disease, Parkinson's disease).
  • a psychiatric disorder e.g., schizophrenia, obsessive compulsive disorder, addiction, depression, anxiety, psychosis
  • an autism spectrum disorder e.g., Fragile X syndrome, Rett syndrome
  • epilepsy e.g., chronic traumatic encephal
  • the neurological condition or disease is any seizure and/or epilepsy related condition or disease wherein PV neurons are implicated.
  • the nucleic acid cassette results in selective expression in PV neurons at a level that is at least 2 fold, at least 5 fold, or at least 7 fold, or at least 10 fold as compared to CAG or EF1 ⁇ or a non-selective regulatory element operably linked to the transgene, as measured by a co-localization assay.
  • the nucleic acid cassette is in an AAV. In some cases, the AAV is AAV9.
  • a method of treating Dravet syndrome comprises contacting a cell with an AAV comprising a transgene that is any one of (i) SCN1A, SCN1B, SCN2B, a DNA binding protein (ii) a functional fragment thereof, or (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the AAV further comprises one or more PV neuron selective regulatory elements or any of the regulatory elements disclosed herein operably linked to the transgene.
  • each of the regulatory elements independently comprises a sequence comprising any one of SEQ ID NOs: 1-32, or any functional fragment or combination thereof, or a sequence comprising at least 80% sequence identity to any one of SEQ ID NOs: 1-32.
  • a method of treating Alzheimer's disease comprises contacting a cell with an AAV comprising a transgene is any one of (i) SCN1A, SCN2B, KV3.1, KV3.3, and STXBP1, (ii) a functional fragment thereof, and (iii) a sequence having at least 80% sequence identity to (i) or (ii).
  • the AAV further comprises one or more PV neuron selective regulatory elements operably linked to the transgene.
  • each of the regulatory elements independently comprises a sequence comprising any one of SEQ ID NOs: 1-32, or any functional fragment or combination thereof, or a sequence comprising at least 80% sequence identity to any one of SEQ ID NOs: 1-32.
  • PV cells can be harvested from a R26-CAG-LSL-Sun1-sfGFP-Myc knockin mouse using affinity purification, e.g., using anti-GFP or anti-Myc antibodies and protein G-coated magnetic beads.
  • PV cells can be enriched by using anti-PV antibody coated beads or affinity purification matrix.
  • Nuclei are then isolated from the PV cells.
  • Nuclear RNA can be purified from the nuclei and converted to cDNA, and amplified with the Nugen Ovation RNA-seq System V2 (Nugen 7102), followed by sequencing using the Illumina HiSeq 2500.
  • Genomic DNA can be purified from nuclei, fragmented, and used to make methylC-seq libraries, which can be sequenced using the Illumina HiSeq 2000.
  • nuclei bound to beads are transposed using Tn5 transposase (Illumina FC-121-1030). After 9-12 cycles of PCR amplification, libraries are sequenced using an Illumina HiSeq 2500.
  • Tn5 transposase Illumina FC-121-1030
  • libraries are sequenced using an Illumina HiSeq 2500.
  • ChIP-seq library nuclei of PV cells are digested to mononucleosomes using micrococcal nuclease, followed by salt extraction of chromatin, and native ChIP and library construction, which can be sequenced on an Illumina HiSeq 2500.
  • sequences are mapped to identify correlations and patterns in hypo-methylation in CG-rich regions, histone modifications, transcriptional factor binding sites, and patterns associated with highly expressed transcriptional factors in PV cells. Overlapping features and correlations from multiple assays and/or libraries described above provide convergent evidence for identifying candidate sequences that are putative PV-selective regulatory elements. Putative PV-selective regulatory elements can be further tested using a co-localization assay as described in Example 5 below.
  • Putative PV-selective regulatory elements can also be tested in B6 PV-Cre mouse (Jackson Laboratory), which is a B6 PV-Cre knock-in mouse that expresses Cre recombinase in parvalbumin-expressing, as described in Example 2 below. After validating PV-selectivity of the regulatory elements, the regulatory elements can be operably linked to a transgene to target expression selectively to PV cells over at least one, two, three, four, five, or more than five non-PV cells.
  • AAV9 vectors containing eGFP operably linked to (i) a control promoter (EF1 ⁇ ); or (ii) a PV-selective RE identified in Example 1 above; or (iii) a PV-selective RE selected SEQ ID NOs: 1-32; and AAV9 vectors containing a Cre dependent tdTomato are co-injected into a B6 PV-Cre mouse (Jackson Labs).
  • PV-Cre is a knock-in mouse that expresses Cre recombinase in parvalbumin-expressing neurons (such as interneurons in the brain and proprioceptive afferent sensory neurons in the dorsal root ganglia), without disrupting endogenous Pvalb expression.
  • mice are infused bilaterally with 1.5 ⁇ L of AAV9 vector (5 12 to 1 13 gc/ml) into the dorsal and ventral hippocampus at a rate of 0.3 ⁇ L/min with a 4 min rest period following injection. Mice are anesthetized for the injection.
  • the animals are placed in a stereotaxic frame (Kopf instruments, USA), using the following coordinates for the dorsal hippocampus (AP ⁇ 2.0 mm, lateral ⁇ 1.5, DV ⁇ 1.4 mm from dura) and the ventral hippocampus (AP ⁇ 3.1 mm, lateral ⁇ 2.8, DV ⁇ 3.8 mm from dura).
  • a Hamilton syringe (model #80308; 10 ⁇ L syringe with corresponding 30 ga blunt tip needle) can be used with the stereotactic micromanipulator, to designate and drill the bur holes.
  • the drill is only used to penetrate the bone.
  • the infusion cannula is lowered into the brain to the depth of the desired location for injection, e.g., injection volume: 1.5 ⁇ L; injection rate: 0.3 ⁇ L/min.
  • injection volume 1.5 ⁇ L
  • injection rate 0.3 ⁇ L/min.
  • the needle Prior to infusion, the needle is allowed to equilibrate for 1 minute. Once delivery is completed, the needle is left for 4 min and then withdrawn over approximately 1 min. Once all infusions are complete, the skin incision is closed with sutures and administered post-surgery analgesics.
  • the treated mice undergo daily health checks for the remainder of the study and are weighed once weekly to monitor body weight.
  • mice are euthanized via isoflurane overdose and perfused with 4% Paraformaldehyde (PFA).
  • PFA Paraformaldehyde
  • a piece of brain tissue containing the hippocampus is extracted and placed in 4% PFA at 4° C. for at least 12 hours.
  • the brain tissue is then dehydrated in 30% sucrose (in phosphate buffered saline) at 4° C. until the tissue sinks to the bottom of the tube.
  • Brain tissue is embedded in Tissue-Tek OCT for sectioning in a cryostat.
  • Sectioned brain tissue is stained for eGFP and tdTomato using standard immunohistochemistry procedures with anti-RFP polyclonal rabbit antibody (Rockland Antibodies and Assay) and anti-eGFP polyclonal chicken antibody (Ayes Labs). Fluorescence microscope imaging is used to visualize the cells. eGFP, or green fluorescence, corresponds to all gene expression. Red fluorescence from tdTomato corresponds to PV+ cells. An overlap of the two fluorescence signals, which can be visualized as yellow or white cells, represents PV+ cells that express the eGFP transgene.
  • AAV9 vectors comprising a PV-selective regulatory element is expected to yield higher number of cells that are eGFP+ and PV+ as compared to the control promoter (EF1 ⁇ ).
  • EF1 ⁇ control promoter
  • fluorescence imaging of cells from mice injected with AAV9s comprising any one of PV-selective REs are expected to show higher number of eGFP+ cells that are also PV+.
  • Selectivity for PV cells can be quantified as percentage of all eGFP+ cells that are also PV+.
  • mice B6(Cg)-Scn1a tm1.1Dsf /J mice were obtained from the Dravet syndrome European Federation via the Jackson Laboratories. These mice contain a Dravet syndrome associated mutation in exon 24 of SCN1A (A to V at position 1783). The mice also contain a floxed exon 24 with wildtype sequence. When not manipulated, this strain of mice expresses two copies of the WT allele of SCN1A. However, upon delivery of an AAV expressing Cre recombinase, any cell targeted by the AAV will switch to expressing one copy of the mutant allele. Upon expression of the mutant SCN1A subunit, mice develop spontaneous seizures within 10 days.
  • FIG. 1 illustrates the frequency of seizures in 12 hour windows over 14 days following treatment.
  • the mice treated with SCN1B showed a trend towards lower seizure frequency compared to the control animals.
  • beta unit of the sodium ion channel e.g, SCN1B
  • SCN1B can contribute to the trafficking and assembly of the sodium ion channel and that increasing the expression of the beta unit selectively in PV neurons can result in increased trafficking and assembly of the Nav1.1 channel, thus leading to a trend towards lower seizure frequency and duration in the mice treated with SCN1B gene therapy.
  • APP/PS1 mice Female APP/PS1 and WT mice bred at PsychoGenics were used in the study.
  • APP/PS1 mice contain human transgenes for both Amyloid Beta Precursor Protein (APP) bearing the Swedish mutation (670 G-T and 671 A-C) and Presenilin 1 (PSEN1) containing an L166P mutation, both under the control of the Thy1 promoter. These mice develop symptoms of Alzheimer's disease, including amyloid plaques and memory defects. Further description of these mice can be found in Radde et al, 2006 (Radde, Rebecca, et al. “A ⁇ 42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology.” EMBO reports 7.9 (2006): 940-946).
  • APP/PS1 mice were used as a model to determine the effect of treatment with SCN1B under the control of a RE on symptoms of Alzheimer's disease.
  • APP/PS1 mice and non-transgenic controls were injected with either a control vector expressing eGFP or a treatment vector expressing SCN1B, both under the control of SEQ ID NO: 32; and implanted with an EET transmitter as in Example 3.
  • Brain activity was assessed over 24 hours at 4 weeks after surgery. Electrocorticogram data was automatically analyzed and power levels in the different frequency bands were compared.
  • FIG. 2 illustrates the high gamma power (50-100 Hz) in non-transgenic controls (WT), APP/PS1, and APP/PS1 mice treated with SCN1B.
  • Increased high gamma power activity is associated with seizures in Alzheimer's patients and epilepsy patients.
  • the APP/PS1 mice showed a higher level of high gamma power activity than the control mice. However, the increase was absent in the treated mice indicating effective treatment with the vector.
  • C57BL/6J (WT) mouse line was used for the PV immunohisotochemical assays.
  • Expression cassettes comprising reporter transgene eGFP operably linked to a regulatory element (SEQ ID NO: 1 or SEQ ID NO: 8) or a CAG promoter in an AAV9 construct.
  • mice Postnatal day 1 C57BL/6J mice were infused via facial vein injection with AA9 vector (1 E 12 to 3 E 12 ) using a 300 U insulin syringe with a 31 G needle.
  • mice were euthanized 21 days post-infusion via overdose of sodium pentobarbital (i.p.) and perfused with heparinized (2.5 IU/ml) saline followed by perfusion with 4% formaldehyde.
  • Brains were removed and subsequently immersion-fixed in 4% formaldehyde for 24-48 hours at 4 degrees Celsius. The brain was then placed into PBS containing 30% sucrose and allowed to sink at 4 degrees Celsius ( ⁇ 2-3 days).
  • mice 4-week-old, C57BL/6 mice were infused via tail vein injection with 60 ⁇ L of AAV9 vector (4.9 13 to 1 14 gc/ml) expressing eGFP.
  • AAV9 vector 4.9 13 to 1 14 gc/ml
  • eGFP eGFP
  • mice were euthanized 21 days post-infusion via isoflurane overdose and whole brains were extracted, washed with PBS and placed into separate 5 ml tubes containing ice cold 4% formaldehyde. Tissue was fixed at 4 degrees Celsius overnight. The following day, the brain was placed into PBS containing 30% sucrose and allowed to sink at 4 degrees Celsius. Upon sinking the individual brain hemispheres were frozen in Tissue-Tek OCT with the midline facing down.
  • Frozen brains were processed for sagittal sections on a cryostat and placed free-floating into PBS. Sections were stained for EGFP and parvalbumin (PV) using standard immunohistochemistry procedures with chicken anti-GFP (Ayes Lab, GFP-1020) and mouse anti-PV (Sigma, P3088).
  • Immunohistochemistry protocol Immunohistochemistry was used to analyze the co-localization of eGFP signal and PV signal using the anti-PV antibody, wherein overlay of the signals exhibited as white or light gray spots in the top panel images (merge), wherein representative overlay was indicated by arrowheads. Overlay of the eGFP and PV fluorescence is indicative of expression in PV cells. Such experiments can be used to determine the selective expression of expression in PV cells.
  • tissue obtained from each mouse were blocked with a Blocking Buffer Solution (comprising 3% BSA, 3% NGS, 0.3% Triton X-100, 0.2% Tween-20 in 1 ⁇ PBS) for 1 hour at room temperature.
  • the tissues were then incubated with primary antibodies in blocking buffer overnight at 4C, washed three times with 1 mL 1 ⁇ PBS, each with 5 minutes interval. Then the tissues were incubated with secondary antibodies in blocking buffer for 1 hour at room temperature, followed by washing three times, each time with 1 mL 1 ⁇ PBS and with 5 minutes interval. The tissues were incubated DAPI (1:1000) in PBS buffer for 5 minute and wash twice with 1 mL 1 ⁇ PBS. Tissues were mounted onto slides, imaged, and analyzed using a fluorescence microscope. Images were taken using a Vectra 3 imaging system (Perkin Elmer) and quantified for co-labeling of eGFP and PV staining using inform-Tissue finder, advanced image analysis software or hand scored. At least 80 GFP positive cells were counted in each panel before determining the percentage of co-localization.
  • FIGS. 3A-3C illustrates the results of the immunohistochemistry experiments performed in pups after systemic AAV9 injections.
  • FIGS. 4A-4C illustrates results of similar immunohistochemistry experiments performed in adult mice following AAV9 injections.
  • FIG. 3A illustrates the overlay of the immunohistochemistry experiments performed in pups after systemic AAV9 injections.
  • FIG. 3B illustrates the quantification of the co-localization of the immunohistochemistry experiments, wherein selectivity for PV cells was measured as percentage of GFP+ cells that were also PV+, as compared to eGFP expression under the control of the CAG promoter.
  • FIG. 4A illustrates the overlay of the immunohistochemistry experiments performed in adult mice after systemic AAV9 injections.
  • FIG. 4B illustrates the quantification of the co-localization of the immunohistochemistry experiments, wherein selectivity for PV cells was measured as percentage of GFP+ cells that were also PV+, as compared to eGFP expression under the control of the EF1 ⁇ .
  • GABAergic neurons constitute about 20% of CNS
  • PV cells constitute about 40% of GABAergic neurons, which means that PV cells make up approximately 8% of all neurons in the CNS.
  • a non-selective regulatory element e.g., CAG, EF1 ⁇ , or a constitutive promoter
  • expression in PV cells above 8% is indicative of increased selectivity in PV cells.
  • AAV9 injections comprising regulatory element SEQ ID NO: 8 resulted in about 60% of cells as PV positive, which was 7.5 times higher than what was expected by the distribution of PV cells.
  • 5A-5F illustrate the fluorescence imaging used for determining co-localization, or selectivity, measured as percentage of eGFP positive cells that were also PV positive and in comparison to the signal of non-selective regulatory element SEQ ID NO: 34.
  • Cells that were positive for a marker appear as white/gray cells in the images.
  • Merge images illustrate the overlap between the corresponding eGFP and anti-PV images.
  • Cells that were positive for both eGFP and PV appear as white/light gray cells in the merge image.
  • FIG. 6 illustrates the quantification of the co-localization analysis, measured as percentage of eGFP+ cells that were also PV+.
  • Treatment of Dravet syndrome and/or symptoms thereof using the expression cassettes described herein can be tested in various mouse lines, such as B6(Cg)-Scn1a tm1.1Dsf /J as described above, Scn1a tm1Kea , and Scn1a-R1470X mouse lines. These mouse lines are established mouse models for Dravet syndrome. Scn1a tm1Kea and Scn1a-R1470X mouse lines do not require CRE recombinase.
  • the Scn1a tm1Kea mouse (available from the Jackson Laboratory; described in Hawkins et al., Scientific Reports, vol. 7: 15327 (2017)) comprises a deletion of the first coding exon of SCN1A.
  • Mice homozygous for the SCN1A knockout allele are characterized by tremors, ataxia, seizures, and die by postnatal day 16. Heterozygous mice on the C57BL/6 background develop spontaneous seizures and die within weeks.
  • Such mouse strain can be used to study safety and efficacy of treatment of epilepsy and Dravet syndrome. See Miller et al., Genes Brain Behav. 2014 February; 13(2):163-72 for additional information.
  • the Scn1a-R1470X mouse is a knock-in mouse carrying a premature stop codon, R1407X, in exon 21 of the SCN1A gene.
  • the same mutation has been identified as a pathogenic mutation in three unrelated SME1 patients.
  • Scn1a RX/RX pups are characterized by recurrent spontaneous seizures at 12 postnatal days, including tonic-clonic and clonic seizures at 12---16 postnatal days, and rhythmic jerking movements and involuntary muscle contraction. See Ogiwara et al., Journal of Neuroscience, May 30, 2007, 27 (22) 5903-5914 for additional information,
  • Dravet mice of each of the mouse strains described above and control mice are injected (e.g., administered by intraperitoneal injection) with AAVs expressing either eGFP or another reporter gene, or an expression cassette comprising one or more PV-selective REs SEQ ID NOs: 1-32) as described herein operably linked to a transgene disclosed herein, such as SCN1A, SCN1B, or SCN2B, or any of SEQ ID NOs: 37-39, or a variant or functional fragment thereof.
  • mice survival is monitored over time. All mice are monitored daily for general health (e.g. weight, hydration, grooming, and mobility) and deaths were recorded. Telemetry implantation can be performed immediately after AAV injections (F20-EET, Data Sciences International). Electrocorticogram data can be recorded and monitored continuously for at least 14 days from 10 days after the surgery. All seizure events can be recorded for at least 14 days following AAV treatment, annotated with date, time start, time stop, duration, and severity score. A reduction in the frequency and/or duration of seizures following treatment with an AAV as described above as compared to the eGFP control or an untreated control is indicative of the efficacy of the gene therapy in reducing the symptoms and/or severity of Dravet syndrome.
  • the expression levels of the transgene can be monitored over time using various PCR and/or sequencing methods to show AAV treatment can result in an increase in gene expression in PV cells.
  • Northern blot analysis and in situ hybridization can also be used to analyze transgene expression in vivo.
  • the level of the protein expressed from the protein can also be monitored after treatment to show an increase in transgene expression correlates with an increase in the corresponding protein in vivo.
  • Protein levels can be assayed using various methods, including, but not limited to, Western blot analysis, immunohistochemistry, immunofluorescence histochemistry, and/or ELISA assays. Formation of functional voltage-gated sodium ion channels can also be assayed using current-clamp analysis.
  • Hyperthermia-induced seizures can be evaluated to compare the wild-type mice and/or untreated Dravet mice with Dravet mice treated with AAV gene therapy comprising an expression cassette described herein (e.g., an expression cassette comprising one or more REs of this disclosure operably linked to a transgene of this disclosure, such as SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, a functional fragment thereof, or a DNA binding protein that modulates an endogenous SCN1A, SNC2A, SNC8A, SCN1B, or SCN2B).
  • an expression cassette described herein e.g., an expression cassette comprising one or more REs of this disclosure operably linked to a transgene of this disclosure, such as SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, a functional fragment thereof, or a DNA binding protein that modulates an endogenous SCN1A, SNC2A, SNC8A, SCN
  • the core body temperature is monitored with a RET-3 rectal temperature probe (Physitemp Instruments, Inc, New Jersey, USA) and controlled by a heat lamp connected to a rodent temperature regulator (TCAT-2DF, Physitemp) reconfigured with a Partlow 1160+controller (West Control Solutions, Brighton, UK).
  • Body temperature is raised 0.5° C. every two minutes until the onset of the first clonic convulsion.
  • Dravet mice treated with an AAV gene therapy are expected to have a higher threshold temperature before the onset of first clonic convulsion and/or have a higher proportion of mice that remain seizure free at the maximum temperature tested.
  • AAV comprising an expression cassette
  • mice can also be administered to mice to determine the safety and efficacy profile of each gene therapy treatment.
  • These preclinical studies can also inform the optimal dose(s) of the gene therapy to use for treating Dravet syndrome.
  • mice Female APP/PS1 and wild-type (WI) mice, which are bred at PsychoGenics and are established mouse model of Alzheimer's disease, can be used to study the safety and efficacy of the compositions described herein in treating Alzheimer's disease, comprising one or more PV-selective REs.
  • APP/PS1 mice is describe above in Example 4.
  • APP/PS1 mice and non-transgenic controls are injected with either a control AAV vector expressing eGFP or a treatment AAV vector comprising one or more PV-selective REs disclosed herein, e.g., SEQ ID NOs: 1-32, operably linked to a transgene that is deficient or impaired in Alzheimer's disease, such as SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, STXBP1, a DNA binding protein that modulates an endogenous gene (e.g., SCN1A, SNC2A, SNC8A, SCN1B, SCN2B, KV3.1, KV3.3, or STXBP1), or any one of SEQ ID NOs: 37-43, or a functional fragment thereof.
  • a control AAV vector expressing eGFP or a treatment AAV vector comprising one or more PV-selective REs disclosed herein, e.g., SEQ ID NOs: 1-32, operably
  • mice survival is monitored over time. All mice are monitored daily for general health (e.g. weight, hydration, grooming, and mobility) and deaths were recorded.
  • mice are also implanted with an EET transmitter as described in Example 3 above. Brain activity can be recorded and monitored over 24 hours for at least 4 weeks after surgery.
  • Electrocorticogram data can be automatically analyzed, and power levels in the different frequency bands (50-100 Hz) can be compared across different groups: WT mice, untreated APP/PS1 mice, and AAV-treated APP/PS1 mice, each treated with an AAV gene therapy as described above.
  • Increased high gamma power activity is associated with seizures in Alzheimer's patients and epilepsy patients.
  • the untreated APP/PS 1 mice are expected to show a higher level of high gamma power activity than the control mice, while this increase is expected to he absent or reduced in the treated mice, indicating an effective treatment with an AAV gene therapy.
  • the expression levels of the transgene can be monitored over time using various PCR and/or sequencing methods to show AAV treatment can result in an increase in endogenous expression of the transgene.
  • Northern blot analysis and in situ hybridization can also be used to analyze gene expression in vivo.
  • the level of the protein expressed from the transgene can also be monitored after treatment to show an increase in gene expression correlates with an increase in protein levels.
  • Protein level can be assayed using various methods, including, but not limited to, Western blot analysis, immunohistochemistry, and/or ELBA assays. Formation of functional voltage-gated sodium or potassium ion channels can also be assayed using current-clamp analysis.
  • AAV comprising an expression cassette
  • mice can also be administered to mice to determine the safety and efficacy profile of each gene therapy treatment.
  • These preclinical studies can also inform the optimal dose(s) of the gene therapy to use for treating Alzheimer's disease.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pain & Pain Management (AREA)
  • Environmental Sciences (AREA)
US16/153,443 2017-04-03 2018-10-05 Tissue selective transgene expression Abandoned US20190024121A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/153,443 US20190024121A1 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762480998P 2017-04-03 2017-04-03
PCT/US2018/025940 WO2018187363A1 (en) 2017-04-03 2018-04-03 Tissue selective transgene expression
US16/153,443 US20190024121A1 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/025940 Continuation WO2018187363A1 (en) 2017-04-03 2018-04-03 Tissue selective transgene expression

Publications (1)

Publication Number Publication Date
US20190024121A1 true US20190024121A1 (en) 2019-01-24

Family

ID=63713399

Family Applications (6)

Application Number Title Priority Date Filing Date
US16/153,420 Active US10287608B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/153,443 Abandoned US20190024121A1 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/153,401 Active US10287607B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/153,433 Active US10519465B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/670,996 Abandoned US20200165628A1 (en) 2017-04-03 2019-10-31 Tissue selective transgene expression
US18/055,968 Pending US20230203531A1 (en) 2017-04-03 2022-11-16 Tissue selective transgene expression

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/153,420 Active US10287608B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression

Family Applications After (4)

Application Number Title Priority Date Filing Date
US16/153,401 Active US10287607B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/153,433 Active US10519465B2 (en) 2017-04-03 2018-10-05 Tissue selective transgene expression
US16/670,996 Abandoned US20200165628A1 (en) 2017-04-03 2019-10-31 Tissue selective transgene expression
US18/055,968 Pending US20230203531A1 (en) 2017-04-03 2022-11-16 Tissue selective transgene expression

Country Status (16)

Country Link
US (6) US10287608B2 (ja)
EP (1) EP3607073A4 (ja)
JP (2) JP7246359B2 (ja)
KR (1) KR102604159B1 (ja)
CN (2) CN117821509A (ja)
AU (1) AU2018250161B2 (ja)
BR (1) BR112019020777A2 (ja)
CA (1) CA3058189A1 (ja)
CL (2) CL2019002805A1 (ja)
CO (1) CO2019011450A2 (ja)
EA (1) EA201992358A1 (ja)
IL (1) IL269767A (ja)
MX (4) MX2019011772A (ja)
SG (1) SG11201909203WA (ja)
TW (3) TW202315946A (ja)
WO (1) WO2018187363A1 (ja)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10538764B2 (en) 2014-06-16 2020-01-21 University Of Southampton Reducing intron retention
US10683503B2 (en) 2017-08-25 2020-06-16 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
US10696969B2 (en) 2014-10-03 2020-06-30 Cold Spring Harbor Laboratory Targeted augmentation of nuclear gene output
US10941405B2 (en) 2015-10-09 2021-03-09 University Of Southampton Modulation of gene expression and screening for deregulated protein expression
US11083745B2 (en) 2015-12-14 2021-08-10 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant mental retardation-5 and Dravet Syndrome
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
US11814622B2 (en) 2020-05-11 2023-11-14 Stoke Therapeutics, Inc. OPA1 antisense oligomers for treatment of conditions and diseases

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201909203WA (en) 2017-04-03 2019-11-28 Encoded Therapeutics Inc Tissue selective transgene expression
CA3096407A1 (en) * 2018-04-09 2019-10-17 Allen Institute Rescuing voltage-gated sodium channel function in inhibitory neurons
CN113966399A (zh) 2018-09-26 2022-01-21 加州理工学院 用于靶向基因疗法的腺相关病毒组合物
CN113966400A (zh) * 2019-02-05 2022-01-21 博德研究所 用于使神经元细胞兴奋性正常化和治疗德拉韦综合征的中间神经元特异性治疗剂
CN113710693A (zh) * 2019-02-25 2021-11-26 马萨诸塞大学 Dna结合结构域反式激活因子及其用途
WO2020198017A1 (en) * 2019-03-22 2020-10-01 Encoded Therapeutics, Inc. Multiplexing regulatory elements to identify cell-type specific regulatory elements
SG11202113048SA (en) * 2019-05-29 2021-12-30 Encoded Therapeutics Inc Compositions and methods for selective gene regulation
EP4179098A1 (en) * 2020-07-08 2023-05-17 Baylor College Of Medicine Gene therapy for stxbp1 encephalopathy
KR20230037586A (ko) * 2020-07-09 2023-03-16 가부시키가이샤 모달리스 Mapt 유전자를 표적으로 하는 알츠하이머병의 치료 방법
AU2022378524A1 (en) * 2021-10-28 2024-05-02 UCB Biopharma SRL Nucleic acid constructs, viral vectors and viral particles

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150353917A1 (en) * 2014-06-05 2015-12-10 Sangamo Biosciences, Inc. Methods and compositions for nuclease design

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7405A (en) * 1850-05-28 John weidman
JPH10501686A (ja) 1994-04-13 1998-02-17 ザ ロックフェラー ユニヴァーシティ 神経系の細胞へのdnaのaav仲介送達
WO1998046273A2 (en) 1997-04-17 1998-10-22 Paola Leone Delivery system for gene therapy to the brain
CA2205076A1 (en) 1997-05-14 1998-11-14 Jim Hu Episomal expression cassettes for gene therapy
AU746454B2 (en) 1998-03-02 2002-05-02 Massachusetts Institute Of Technology Poly zinc finger proteins with improved linkers
US6303370B1 (en) 1998-03-24 2001-10-16 Mayo Foundation For Medical Education And Research Tissue-specific regulatory elements
CA2246005A1 (en) 1998-10-01 2000-04-01 Hsc Research And Development Limited Partnership Hybrid genes for gene therapy in erythroid cells
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US6649371B1 (en) 1999-06-11 2003-11-18 Neurosearch A/S Potassium channel KCNQ5 and sequences encoding the same
DE1252330T1 (de) * 1999-11-26 2003-11-27 Univ Montreal Mcgill Loci der idiopathischen epilepsie, mutationen derselben und verfahren zu deren verwendung zur feststellung, prognose und behandlung von epilepsie
AU776576B2 (en) 1999-12-06 2004-09-16 Sangamo Biosciences, Inc. Methods of using randomized libraries of zinc finger proteins for the identification of gene function
WO2001094414A2 (en) 2000-06-07 2001-12-13 Ortho Mcneil Pharmaceutical, Inc. THE HUMAN VOLTAGE GATED SODIUM CHANNEL β-1A SUBUNIT AND METHODS OF USE
AUPR492201A0 (en) 2001-05-10 2001-06-07 Bionomics Limited Novel mutation
EP1402043A1 (en) 2001-07-03 2004-03-31 Institut National De La Sante Et De La Recherche Medicale (Inserm) Methods of administering vectors to synaptically connected neurons
US6998118B2 (en) 2001-12-21 2006-02-14 The Salk Institute For Biological Studies Targeted retrograde gene delivery for neuronal protection
CA2474751A1 (en) 2002-02-25 2003-09-04 Vanderbilt University Expression system for human brain-specific voltage-gated sodium channel, type 1
US7361635B2 (en) 2002-08-29 2008-04-22 Sangamo Biosciences, Inc. Simultaneous modulation of multiple genes
ATE446371T1 (de) 2003-01-28 2009-11-15 Ludwig Aigner Verwendung von regulatorischen sequenzen zur spezifischen, transienten expression in neuronal determinierten zellen
US20040258666A1 (en) 2003-05-01 2004-12-23 Passini Marco A. Gene therapy for neurometabolic disorders
US7094600B2 (en) 2003-06-26 2006-08-22 The Research Foundation Of State University Of New York Screen for sodium channel modulators
JP2007532639A (ja) 2004-04-14 2007-11-15 エージェンシー フォー サイエンス,テクノロジー アンド リサーチ 神経細胞への遺伝子送達方法
TWI293307B (en) 2004-09-30 2008-02-11 Ind Tech Res Inst A liver-specific chimeric regulatory sequence and use thereof
EP2021499A4 (en) * 2005-12-16 2010-02-17 Univ Leland Stanford Junior FUNCTIONAL ARRAYS FOR THE IDENTIFICATION OF ELEMENTS FOR GENE PRESSION REGULATION WITH HIGH THROUGHPUT
US20070161031A1 (en) 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
WO2007084957A2 (en) 2006-01-20 2007-07-26 University Of California, San Francisco Transplantation of neural cells
EP2097538A4 (en) 2006-12-07 2011-11-30 Switchgear Genomics TRANSCRIPTION REAGULATION ELEMENTS OF BIOLOGICAL PATHS, TOOLS AND METHODS
EP2019143A1 (en) 2007-07-23 2009-01-28 Genethon CNS gene delivery using peripheral administration of AAV vectors
US20100297674A1 (en) 2008-01-22 2010-11-25 Chromocell Corporation NOVEL CELL LINES EXPRESSING NaV AND METHODS USING THEM
US20110165129A1 (en) 2008-05-06 2011-07-07 Arnold Kriegstein Ameliorating Nervous Systems Disorders
US9267151B2 (en) * 2008-08-20 2016-02-23 Brainco Biopharma, S.L. STXBP1 overexpressing mouse and its uses in screening of treatments for neuropsychiatric illness
WO2010037143A1 (en) 2008-09-29 2010-04-01 The University Of Montana Vectors and methods of treating brain seizures
US8815779B2 (en) 2009-09-16 2014-08-26 SwitchGear Genomics, Inc. Transcription biomarkers of biological responses and methods
US20110135611A1 (en) 2009-12-03 2011-06-09 The J. David Gladstone Institutes Methods for treating apolipoprotein e4-associated disorders
US9315825B2 (en) 2010-03-29 2016-04-19 The Trustees Of The University Of Pennsylvania Pharmacologically induced transgene ablation system
CA2833908C (en) 2010-04-23 2021-02-09 University Of Massachusetts Cns targeting aav vectors and methods of use thereof
US8927514B2 (en) 2010-04-30 2015-01-06 City Of Hope Recombinant adeno-associated vectors for targeted treatment
LT2566972T (lt) 2010-05-03 2020-03-10 Sangamo Therapeutics, Inc. Kompozicijos, skirtos cinko pirštu modulių susiejimui
PL2585596T3 (pl) 2010-06-23 2021-06-28 Curna, Inc. Leczenie chorób związanych z podjednostką alfa kanału sodowego bramkowanego napięciem (SCNA) poprzez hamowanie naturalnego transkryptu antysensownego SCNA
JP5704361B2 (ja) 2010-10-27 2015-04-22 学校法人自治医科大学 神経系細胞への遺伝子導入のためのアデノ随伴ウイルスビリオン
EP2635686B1 (en) 2010-11-05 2018-03-28 The Regents of The University of California Neuronal specific targeting of caveolin expression to restore synaptic signaling and improve cognitive function in the neurodegenerative brain and motor function in spinal cord
WO2012065143A1 (en) * 2010-11-12 2012-05-18 The General Hospital Corporation Polycomb-associated non-coding rnas
EP2655621B1 (en) * 2010-12-20 2018-05-23 The General Hospital Corporation Polycomb-associated non-coding rnas
WO2012112578A2 (en) 2011-02-14 2012-08-23 The Children's Hospital Of Philadelphia Improved aav8 vector with enhanced functional activity and methods of use thereof
WO2013123503A1 (en) 2012-02-17 2013-08-22 The Children's Hospital Of Philadelphia Aav vector compositions and methods for gene transfer to cells, organs and tissues
US9284575B2 (en) 2012-03-06 2016-03-15 Duke University Synthetic regulation of gene expression
WO2013155222A2 (en) 2012-04-10 2013-10-17 The Regents Of The University Of California Brain-specific enhancers for cell-based therapy
KR102527259B1 (ko) 2013-03-15 2023-04-27 더 칠드런스 호스피탈 오브 필라델피아 스터퍼/필러 폴리누클레오티드 서열을 포함하는 벡터 및 사용 방법
BR112015028605A8 (pt) 2013-05-15 2019-12-24 Univ Minnesota uso de uma composição e uso de um imunossupressor e uma composição
JP6594891B2 (ja) 2014-03-18 2019-10-23 サンガモ セラピューティクス, インコーポレイテッド ジンクフィンガータンパク質発現を調節するための方法および組成物
US11020443B2 (en) 2015-04-23 2021-06-01 University Of Massachusetts Modulation of AAV vector transgene expression
CN104846015B (zh) 2015-05-27 2018-03-27 深圳先进技术研究院 特异性兴奋伏隔核中的gaba能神经元的组合物及其在改善精神分裂症异样行为中的应用
WO2017075335A1 (en) 2015-10-28 2017-05-04 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
MX2018004755A (es) 2015-10-29 2018-12-19 Voyager Therapeutics Inc Entrega de polinucleotidos dirigidos al sistema nervioso central.
SG11201909203WA (en) 2017-04-03 2019-11-28 Encoded Therapeutics Inc Tissue selective transgene expression

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150353917A1 (en) * 2014-06-05 2015-12-10 Sangamo Biosciences, Inc. Methods and compositions for nuclease design

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10538764B2 (en) 2014-06-16 2020-01-21 University Of Southampton Reducing intron retention
US11891605B2 (en) 2014-06-16 2024-02-06 University Of Southampton Reducing intron retention
US10696969B2 (en) 2014-10-03 2020-06-30 Cold Spring Harbor Laboratory Targeted augmentation of nuclear gene output
US10941405B2 (en) 2015-10-09 2021-03-09 University Of Southampton Modulation of gene expression and screening for deregulated protein expression
US11702660B2 (en) 2015-10-09 2023-07-18 University Of Southampton Modulation of gene expression and screening for deregulated protein expression
US11083745B2 (en) 2015-12-14 2021-08-10 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant mental retardation-5 and Dravet Syndrome
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
US10683503B2 (en) 2017-08-25 2020-06-16 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
US10913947B2 (en) 2017-08-25 2021-02-09 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
US11873490B2 (en) 2017-08-25 2024-01-16 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
US11814622B2 (en) 2020-05-11 2023-11-14 Stoke Therapeutics, Inc. OPA1 antisense oligomers for treatment of conditions and diseases

Also Published As

Publication number Publication date
WO2018187363A1 (en) 2018-10-11
EP3607073A1 (en) 2020-02-12
BR112019020777A2 (pt) 2020-04-28
JP7477675B2 (ja) 2024-05-01
US20190024118A1 (en) 2019-01-24
MX2022015603A (es) 2023-02-09
TWI808079B (zh) 2023-07-11
AU2018250161B2 (en) 2024-04-04
CL2019002805A1 (es) 2020-03-06
JP2023072039A (ja) 2023-05-23
US10287607B2 (en) 2019-05-14
CN117821509A (zh) 2024-04-05
IL269767A (en) 2019-11-28
CL2023000294A1 (es) 2023-08-25
TW202315946A (zh) 2023-04-16
EA201992358A1 (ru) 2020-03-24
MX2022015605A (es) 2023-02-09
EP3607073A4 (en) 2020-12-30
JP7246359B2 (ja) 2023-03-27
MX2022015604A (es) 2023-02-09
US20190024119A1 (en) 2019-01-24
US10519465B2 (en) 2019-12-31
SG11201909203WA (en) 2019-11-28
US20230203531A1 (en) 2023-06-29
CA3058189A1 (en) 2018-10-11
TW201842185A (zh) 2018-12-01
KR20190137126A (ko) 2019-12-10
TW202315945A (zh) 2023-04-16
MX2019011772A (es) 2020-01-09
US10287608B2 (en) 2019-05-14
CO2019011450A2 (es) 2019-10-31
US20190024120A1 (en) 2019-01-24
AU2018250161A1 (en) 2019-10-17
CN110730823B (zh) 2023-12-29
JP2020515292A (ja) 2020-05-28
US20200165628A1 (en) 2020-05-28
CN110730823A (zh) 2020-01-24
KR102604159B1 (ko) 2023-11-20

Similar Documents

Publication Publication Date Title
US10287607B2 (en) Tissue selective transgene expression
US20200397917A1 (en) Engineered dna binding proteins
TWI835761B (zh) 工程化dna結合蛋白
US20230365652A1 (en) Nucleic Acid Constructs, Viral Vectors and Viral Particles
JP2024069591A (ja) 操作されたdna結合タンパク質
CA3234666A1 (en) Nucleic acid constructs, viral vectors and viral particles
JP2023518809A (ja) アデノ随伴ウイルスの改変方法及び分離方法

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: ENCODED GENOMICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TAGLIATELA, STEPHANIE;YOUNG, ANDREW;CHEN, SZU-YING;AND OTHERS;REEL/FRAME:048402/0792

Effective date: 20180403

Owner name: ENCODED THERAPEUTICS, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:ENCODED GENOMICS, INC.;REEL/FRAME:048410/0988

Effective date: 20180503

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION