US20230381340A1 - Aav-mediated delivery of atp1a3 genes to central nervous system - Google Patents

Aav-mediated delivery of atp1a3 genes to central nervous system Download PDF

Info

Publication number
US20230381340A1
US20230381340A1 US18/449,346 US202318449346A US2023381340A1 US 20230381340 A1 US20230381340 A1 US 20230381340A1 US 202318449346 A US202318449346 A US 202318449346A US 2023381340 A1 US2023381340 A1 US 2023381340A1
Authority
US
United States
Prior art keywords
atp1a3
sequence
aav9
promoter
aav
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.)
Pending
Application number
US18/449,346
Inventor
Simon Frost
Natalia Morsci
Neil Hackett
Dolan Sondhi
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.)
Alternating Hemiplegia Of Childhood Foundation
Cure Ahc Inc
Hope For Annabel
Original Assignee
Alternating Hemiplegia Of Childhood Foundation
Cure Ahc Inc
Hope For Annabel
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 Alternating Hemiplegia Of Childhood Foundation, Cure Ahc Inc, Hope For Annabel filed Critical Alternating Hemiplegia Of Childhood Foundation
Priority to US18/449,346 priority Critical patent/US20230381340A1/en
Publication of US20230381340A1 publication Critical patent/US20230381340A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • 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
    • A61K48/0075Medicinal 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 delivery route, e.g. oral, subcutaneous
    • 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
    • C12N15/63Introduction 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; 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; CARE OF BIRDS, FISHES, INSECTS; 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
    • 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

Definitions

  • the present invention relates to the delivery and expression of ATP1A3 genes in the central nervous system.
  • Alternating Hemiplegia of Childhood is a serious and rare neurodevelopmental disorder, affecting approximately 1 in 1,000,000 children.
  • AHC is caused by de novo loss-of-function mutations in the ATP1A3 gene (Heinzen et al., 2012; Rosewich et al., 2012).
  • the gene encodes the ⁇ 3 subunit of the Na+, K+ ATPase, one of two pumps that establish and maintain electrochemical gradients required for the propagation of action potentials in neurons.
  • AHC-associated mutations do not affect expression of ATP1A3 gene or levels of its protein, but significantly reduce ⁇ 3 protein functionality (Heinzen et al., 2012; Li et al., 2015).
  • ATP1A3 gene is expressed in the central nervous system (CNS) and heart of all mammals studied. Brain expression of ATP1A3 is enriched in high frequency GABAergic interneurons of the cerebellum, pons, basal ganglia, thalamus, cortex and hippocampus (B ⁇ ttger et al., 2011; Hieber et al., 1991; McGrail et al., 1991; Pietrini et al., 1992; Richards et al., 2007).
  • Clinical symptoms of cerebellar, hippocampal, basal ganglia, and motor cortex dysfunction largely correlate with persistent symptoms of AHC. In line with clinical symptoms, brains of AHC patients show progressive frontal dominant cerebral, severe hippocampal, and diffuse cerebellar atrophy (Sasaki et al., 2017). Paroxysmal symptoms of AHC likely originate in Purkinje cells and spread through the afferent networks of the cerebellum by spreading depolarization (“spreading depression”).
  • AHC symptoms appear before 18 months of age, and often shortly after birth. AHC causes severe morbidity and increased mortality. Patients often suffer severe neurodevelopmental impairments, episodes of paralysis, severe dystonia, ataxia, swallowing problems, epilepsy, status epilepticus, sudden unexpected death, and episodes of developmental regression (Mikati et al., 2000; Panagiotakaki et al., 2015). Currently, there is no effective on-target treatment, although certain off-target drugs like Flunarizine have been reported to have some limited effect on certain symptoms.
  • mutations in ATP1A3 gene are associated with at least eight other distinct, but clinically overlapping neurological disorders (Dard et al., 2015; de Carvalho Aguiar et al., 2004; Demos et al., 2014; Heinzen et al., 2014; Paciorkowski et al., 2015; Smedemark-Margulies et al., 2016; Sweadner et al., 2016; Sweney et al., 2015; Yano et al., 2017).
  • the clinical symptoms of these diseases partly intersect and range from mild to severe.
  • transgenic vector for transducing cells of the central nervous system of a mammal and transgenically expressing a protein in the central nervous system of the mammal.
  • the transgenic vector comprises a virus-derived vector, a nucleic acid sequence encoding the protein, and an endogenous ATP1A3 promoter (P ATP1A3 ) sequence.
  • compositions comprising a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3) protein, in a form compatible with administration into the central nervous system.
  • AAV adeno-associated virus
  • Another aspect of the invention relates to a method for treating a subject having a neurological disorder associated with mutations in the ATP1A3 gene.
  • the method comprises administering into the central nervous system (CNS) of the subject a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 protein, thereby restoring some degree of neurological function in said subject.
  • CNS central nervous system
  • AAV adeno-associated virus
  • Another aspect of the invention relates to a method for delivering to the central nervous system of a mammal, a ATP1A3 DNA that is expressed in a cell of the central nervous system.
  • the method comprises administering into the central nervous system of the mammalian a recombinant AAV vector comprising a nucleic acid sequence encoding a ATP1A3 protein, wherein said vector transduces the cell.
  • FIG. 1 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 2 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum, as a zoomed image.
  • FIG. 3 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse brain at dorsal hippocampus level. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 4 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse brain at dorsal hippocampus level, as a zoomed image.
  • FIG. 5 depicts ATP1A3 (Millipore #06-172-1) staining in the dorsal hippocampus, as zoomed image.
  • FIG. 6 depicts ATP1A3 (Millipore #06-172-1) staining in the parietal cortex, as a zoomed image.
  • FIG. 7 depicts Calbindin (Synaptic Systems #214006) staining in a mouse cerebellum.
  • FIG. 8 depicts Calbindin (Synaptic Systems #214006) staining in a mouse cerebellum, as a zoomed image.
  • FIG. 9 depicts GAD67 (Synaptic Systems #198 006) staining in a mouse brain at dorsal hippocampus level.
  • FIG. 10 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol, as a zoomed image over the dorsal hippocampus.
  • FIG. 11 depicts GAD67 (Millipore #MAB5406) staining in a mouse brain at dorsal hippocampus level.
  • FIG. 12 depicts: GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol.
  • Zoomed image over the dorsal hippocampus showing good detection of GABAergic neurons in the hippocampus and in the cortex.
  • FIG. 13 depicts mCherry staining in a mouse cerebellum.
  • FIG. 14 depicts the mouse cerebellum anatomy.
  • FIG. 15 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition, as zoomed images.
  • FIG. 16 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition, as zoomed images.
  • FIG. 17 depicts histograms of the mean intensity of ATP1A3 in the 4 experimental groups. Histograms are well superposed.
  • FIG. 18 depicts histograms of the mean intensity of ATP1A3 in the 4 experimental groups with the gate for highest fluorescence levels.
  • FIG. 19 depicts an example of Purkinje cell detection. Contours of detected cells are shown.
  • FIG. 20 depicts an example of Purkinje cell detection showing cells with higher ATP1A3 levels. Contours of detected cells are shown in dark shading, and cells in gate are shown in light shading.
  • FIG. 21 depicts an example of Purkinje cell detection showing cells with higher ATP1A3 levels. Contours of detected cells are shown in dark shading, and cells in gate are shown in light shading.
  • FIG. 22 depicts graphical representations of mean ATP1A3 intensity in Purkinje Neurons and percentage of Purkinje neurons with high ATP1A3 intensity.
  • the left graft shows mean intensity in Purkinje Neurons, and the right graph shows the percentage of Purkinje neurons with High ATP1A3 intensity.
  • FIG. 23 depicts graphical representations of mean ATP1A3 intensity in cerebellar cells other than Purkinje neurons.
  • FIG. 24 depicts a histogram of the mean intensity of mCherry in the mCherry group and animals from ATP1A3 infected groups, with gate for highest fluorescence levels.
  • FIG. 25 depicts an example of Purkinje cell detection showing cells with higher mCherry levels (cells in Gate named Highest). Contours of detected cells are shown in black, and cells in gate are shown in dark shading.
  • FIG. 26 depicts graphical representations of mean mCherry intensity in Purkinje Neurons and the percentage of Purkinje neurons with high mCherry intensity.
  • the left graft shows the mean intensity in Purkinje Neurons.
  • the right graph shows the percentage of Purkinje neurons with High mCherry intensity.
  • FIG. 27 depicts graphical representations of mean mCherry intensity in cerebellar cells other than Purkinje neuron.
  • FIG. 28 depicts counts of mCherry positive cells in mCherry infected brainstems.
  • FIG. 29 depicts an example of cells with higher ATP1A3.
  • FIG. 30 depicts quantification of ATP1A3 immunofluorescence.
  • the left graft shows ATP1A3 immunofluorescence at the surrounding of all detected cells.
  • the middle graft shows the percentage of cells detected as higher expressing ATP1A3 after gating.
  • the right graft shows mean fluorescence in cells in highest ATP1A3 gate.
  • transgenic vector for transducing cells of the central nervous system of a mammal and transgenically expressing a protein in the central nervous system of the mammal.
  • the transgenic vector comprises a virus-derived vector, a nucleic acid sequence encoding the protein, and an endogenous ATP1A3 promoter (P ATP1A3 ) sequence.
  • Suitable viral vectors include those neurotrophic viral vectors, including, but not limited to, adeno-associated viral vectors (AAV), herpes simplex viral vectors and lentiviral vectors. More discussions about the herpes simplex viral vectors may be found in U.S. Pat. No. 5,672,344, which is incorporated herein by reference in its entirety.
  • AAV adeno-associated viral vectors
  • herpes simplex viral vectors More discussions about the herpes simplex viral vectors may be found in U.S. Pat. No. 5,672,344, which is incorporated herein by reference in its entirety.
  • the transgenic vector also comprises a nucleic acid sequence encoding a protein (DNA encoding the protein, e.g., cDNA of protein), the transgene.
  • the transgene encodes a biologically active protein which is transgenically expressed in the central nervous system of a mammal.
  • the transgene may be ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3).
  • the transgenes may also be insulin growth factor-1 (IGF-1), calbindin D28, parvalbumin, HIF1-alpha, SIRT-2, VEGF, SMN-1, SMN-2, CNTF (Ciliary neurotrophic factor), sonic hedgehog (shh), erythropoietin (EPO), lysyl oxidase (LOX), progranulin, prolactin, ghrelin, neuroserpin, or placenta lactogen.
  • the protein-coding nucleic acid may be an DNA sequence, an “exogenous DNA” that is exogenous to both AAV and to the target cell.
  • the DNA may be synthetic DNA, complementary DNA, genomic DNA, or a combination thereof.
  • the DNA may be of any sequence or length, provided that it may be incorporated into the vector and delivered to target cells. Typically, because of the packaging limitations of AAV, the exogenous DNA will have a length of up to 4,400 bases.
  • transgenic vector More than one transgene can be delivered by the transgenic vector, e.g., via more than one virus-derived vector.
  • the transgenic vector comprises multiple AAV vectors and each AAV vector comprises a transgene operably linked to a promoter.
  • the protein-coding nucleic acid is operably linked to a promoter sequence to enable the expression of the trangene from a single AAV vector.
  • the protein-coding nucleic acid shall be operably linked to a promoter sequence so that when the promoter is activated, the coding sequence is transcribed.
  • the protein-coding nucleic acid is operably linked if the linkage does not cause an error in the reading of the downstream sequence. To be “operably linked,” it is not necessary that two sequences be immediately adjacent to one another.
  • the promoter sequence drives transcription of the transgene and defines the cellular specificity of its expression.
  • the promoter sequence is an endogenous ATP1A3 promoter (P ATP1A3 ) sequence.
  • the P ATP1A3 promoter comprises a mouse promoter sequence, SEQ ID NO: 1.
  • the P ATP1A3 promoter comprises a homologous human sequence.
  • the transgenic vector may further comprise one or more nucleic acid regulatory sequence, linked directly or indirectly to the protein-coding nucleic acid sequence.
  • a promoter specific to the gene may be incorporated into the transgenic vector.
  • the nucleic acid regulatory sequence comprises a sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene.
  • the nucleic acid regulatory sequence comprises a 3′-UTR sequence that contains a polyadenylation sequence.
  • the transgenic vector comprises an AAV vector (e.g., a recombinant AAV vector), a P ATP1A3 promoter, and a mCherry transgene.
  • AAV vector e.g., a recombinant AAV vector
  • P ATP1A3 promoter e.g., a P ATP1A3 promoter
  • mCherry transgene e.g., a recombinant AAV vector
  • the transgenic vector comprises AAV9 vector (e.g., recombinant AAV9 vector), a P ATP1A3 promoter, a mCherry transgene, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • AAV9 vector e.g., recombinant AAV9 vector
  • P ATP1A3 promoter e.g., recombinant AAV9 vector
  • a mCherry transgene e.g., a sequence to regulate ribosome binding and/or translation efficiency
  • a 3′-UTR sequence that contains a polyadenylation sequence.
  • the transgenic vector comprises SEQ ID NO: 2.
  • the transgenic vector comprises an AAV vector (e.g., a recombinant AAV vector), a P ATP1A3 promoter, and a ATP1A3 transgene.
  • AAV vector e.g., a recombinant AAV vector
  • P ATP1A3 promoter e.g., a P ATP1A3 promoter
  • ATP1A3 transgene e.g., ATP1A3 transgene
  • the transgenic vector comprises AAV9 vector (e.g., recombinant AAV9 vector), a P ATP1A3 promoter, a ATP1A3 transgene, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • AAV9 vector e.g., recombinant AAV9 vector
  • P ATP1A3 promoter e.g., recombinant AAV9 vector
  • a ATP1A3 transgene e.g., a sequence to regulate ribosome binding and/or translation efficiency
  • a 3′-UTR sequence that contains a polyadenylation sequence.
  • the transgenic vector comprises SEQ ID NO: 3.
  • compositions and methods for intracisternal or intrathecal administration of recombinant AAV comprising a gene that affects neurological function are also disclosed herein.
  • the embodiments of the invention illustrate that some neurological disorders associated with loss-of-function mutations of ATP1A3 genes (e.g., alternating hemiplegia of childhood, “AHC”) can be better addressed with a more targeted delivery of the gene therapy.
  • AHC alternating hemiplegia of childhood
  • the phenotype of an AHC mouse model has been rescued by transgenesis.
  • Inclusion of extra-chromosomal copies of ATP1A3 by transgenesis has been shown to be sufficient to increase Na+, K+ ATPase activity in the brain and confer significant phenotypic improvements compared to non-transgenic mice with the same AHC associated mutation.
  • compositions comprising a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3) protein, in a form compatible with administration into the central nervous system.
  • AAV adeno-associated virus
  • the composition contains an adeno-associated virus (AAV) vector.
  • AAV adeno-associated virus
  • the vector may be a derivative of the adeno-associated virus, into which exogenous DNA is introduced.
  • AAV vector is used herein as a carrier to deliver ATP1A3 cDNA.
  • AAV is a replication-defective, non-enveloped parvovirus, present in humans and some other primate species but not associated with any pathology.
  • Recombinant AAV vectors are used for the development of human therapeutics because they can infect quiescent non-dividing cells (like neurons), mediate long-term gene expression, and, unlike retroviruses, persist in an extrachromosomal state without integrating into the genome of the host cell. Since AAV does not integrate into the host genome, the use of this viral vector mitigates risks associated with unintended off-target effects of genomic editing (Schaefer et al., 2017; Zhang et al., 2015).
  • AAV vectors are derived from single-stranded (ss) DNA parvoviruses that are nonpathogenic for mammals. Briefly, recombinant AAV-based vectors have two open reading frames, the rep and cap viral genes that account for 96% of the viral genome removed, leaving the two flanking 145-basepair (bp) inverted terminal repeats (ITRs), which are used to initiate viral DNA replication, packaging and integration. These ITRs allow for synthesis of the complementary DNA strand. Rep and Cap are translated to produce multiple distinct proteins. When constructing an AAV transfer plasmid, the transgene is placed between the two ITRs, and Rep and Cap are supplied in trans. In the absence of helper virus, wild-type AAV is typically maintained episomally.
  • a single AAV particle can accommodate up to 5 kb of ssDNA, therefore leaving about 4.5 kb for a transgene and regulatory elements, which is typically sufficient.
  • trans-splicing systems as described, for example, in U.S. Pat. No. 6,544,785, which is incorporated herein by reference in its entirety, may nearly double this limit.
  • small (4.8 kb) ssDNA AAV genome having ITR sequences flanking the insert are of ITR2 serotype.
  • AAV vector of any serotype can be used.
  • Particular useful serotypes are those that are particularly suitable for nervous cells, e.g., AAV1, AAV2, AAV4, AAV5, AAV8, AAV9, AAV-PHP.B, or AAVrh10.
  • Several of these AAV serotypes have been utilized in clinical Phase I and II studies for diseases that require CNS delivery (reviewed in (Naso et al., 2017)).
  • Initial testing of AAV2-based RPE65 delivery has been done in canine culture.
  • Other serotype besides those listed herein can be used.
  • pseudotyped AAV vectors may also be used herein.
  • Pseudotyped AAV vectors are those which contain the genome of one AAV serotype in the capsid of a second AAV serotype; for example, an AAV vector that contains the AAV2 capsid and the AAV1 genome or an AAV vector that contains the AAV5 capsid and the AAV 2 genome.
  • AAV-based gene therapy targeted to cells of the CNS has been described in U.S. Pat. Nos. 6,180,613 and 6,503,888, which are incorporated by reference in their entirety.
  • Exemplary AAV vectors are recombinant AAV vectors encoding human protein.
  • the AAV vector is AAV9 vector.
  • AAV9 vector is used to deliver a rescue gene to the CNS neurons (e.g., in a mammal, such as mouse, primate, human).
  • Cell-specificity of AAV infection is determined by the capsid serotype.
  • AAV serotype 9 (AAV9) has natural tropism for neurons and was successfully applied to rescue adult mouse models of diseases such as mucopolysaccharidosis (Fu et al., 2010; Hinderer et al., 2016; Watson et al., 2006) and diabetic neuropathy (Homs et al., 2014).
  • AAV9 Injection of AAV9 into cerebrospinal fluid has been highly efficient in targeting the CNS in adult mice (Bey et al., 2017; Fu et al., 2010) and pigs (Federici et al., 2012).
  • AAV9 vectors showed preferentially higher selectivity of neuronal transduction in adult non-human primate compared to other serotypes (El-Shamayleh et al., 2017).
  • the gene therapy is conducted in various mouse models (that phenocopy many clinical symptoms of AHC) by injecting ATP1A3 DNA-carrying AAV9 capsids into cerebrospinal fluid (via injection into cisterna magna), using a similar method used to rescue adult mouse models of Mucopolysaccharidosis (Hinderer et al., 2016; Watson et al., 2006; DiRosario et al., 2010) and diabetic neuropathy (J. Homs et al., 2014).
  • the ATP1A3-coding nucleic acid may be a DNA sequence, an “exogenous DNA” that is exogenous to both AAV and to the target cell.
  • DNA delivered to that subject may be considered exogenous even though it is identical to a gene native to that subject's species, provided it differs in the regulatory or coding region from the cognate gene of the subject to whom it is delivered, and therefore encodes a different gene product or is expressed to a different degree and/or in different cells, under at least some conditions.
  • the DNA may be synthetic DNA, complementary DNA, genomic DNA, or a combination thereof.
  • the DNA may be of any sequence or length, provided that it may be incorporated into the vector and delivered to target cells.
  • the exogenous DNA will have a length of about 10-5,000 bases, e.g., 100 to 4,000 bases.
  • the size of a typical ATP1A3 cDNA ( ⁇ 3 kb) fits within the carrying capacity of AAV vectors (4.4 kb).
  • a mouse ATP1A3 cDNA sequence is used in ATP1A3-AAV9 vector for gene therapy used in mouse for compatibility.
  • a sequence of the most common transcript isoform of mouse ATP1A3 gene NCBI reference sequence NM_001290469.1, may be used.
  • only the ORF portion of the mRNA is included: from ATG start to TGA stop codons (including the latter).
  • a human ATP1A3 cDNA sequence is used in ATP1A3-AAV9 vector for gene therapy used in human for compatibility.
  • a sequence of the most abundant transcript of human ATP1A3 gene NCBI reference sequence NM_152296.4 (SEQ ID NO: 6), may be used.
  • no tags are added to its coding sequence in order not to interfere with transgenic ATP1A3 protein translation, folding, transport and function.
  • the composition further comprises one or more nucleic acid regulatory sequences, linked directly or indirectly to the ATP1A3-coding nucleic acid sequence.
  • the nucleic acid regulatory sequence comprises a promoter sequence that renders the expression of the ATP1A3-coding nucleic acid sequence CNS-specific.
  • the promoters have the ability to drive the ATP1A3 expression in target areas in central nervous system.
  • the promoter sequence can be neuron-specific or glia-specific.
  • a promoter specific to the gene is incorporated into the AAV construct.
  • Inclusion of gene-specific promoters in recombinant AAVs for gene delivery provides the desired specificity for neuronal transduction and expression within particular target cells in the brain that is not possible with promoters with non-specific expression profile, like CMV (Watakabe et al., 2015).
  • the use of specific promoters to restrict transgene expression was utilized successfully in several gene delivery studies on non-human primates.
  • a 300 bp TH promoter in AAV vector was used to drive expression in dopaminergic neurons of macaques, achieving a specificity of 95% (Stauffer et al., 2016).
  • short 530 bp enhancer sequence Dlx was used to drive GFP expression in GABAergic neurons of marmosets, achieving a specificity of 93% (Dimidschstein et al., 2016).
  • lkb-long L7/Pcp2 promoter was used to drive ChR2 expression in Purkinje neurons with 96% selectivity (El-Shamayleh et al., 2017).
  • the promoter sequence comprises an endogenous ATP1A3 promoter (P ATP1A3 ); a human neuron-specific promoter with a strong bias towards inhibitory neuron (P hSyn ); or an artificial composite promoter comprising CMV enhancer, chicken 3-actin promoter, and MVM intron (P CBh ).
  • an endogenous ATP1A3 promoter is employed.
  • a pig ATP1A3 promoter is employed.
  • a mouse ATP1A3 promoter is employed (see, e.g., SEQ ID NO: 1).
  • a primate ATP1A3 promoter is employed.
  • a human ATP1A3 promoter is employed.
  • the promoter sequence comprises an endogenous ATP1A3 promoter (P ATP1A3 ) consisting of ⁇ 407/+143 of mouse genomic sequence where “/” indicates the transcription start site.
  • Mouse ATP1A3 ⁇ 407/+143 promoter sequence is based on minimal pig and rat ATP1A3 promoters that were sufficient for neuronal expression in vivo (Bierie et al., 2005; Henriksen et al., 2013; Pathak et al., 1994). To avoid redundancy in having two ribosome binding sites in the constructs, the promoter is shortened to +143 to exclude endogenous Kozak sequence immediately adjacent to the start codon.
  • This minor shortening may not have a significant negative effect on transgene expression, because ⁇ 210/+77 rat sequence of ATP1A3 promoter was sufficient to drive brain-specific transgene expression in mice in vivo (Pathak et al., 1994).
  • the promoter sequence comprises 488 bp-long promoter of porcine ATP1A3 closely homologous to human, which was sufficient to drive GFP expression in Zebrafish brain and spinal cord (Henriksen et al., 2013). Because porcine ATP1A3 promoter was sufficient to drive expression in evolutionarily-distant Zebrafish, it may be sufficient to drive expression on closer-related mammal, like the mouse. In total, 550 nt promoter+3 kb cDNA+133nt rabbit beta globin polyA sequence totals 3,683nt and thus fits within the 4.4 kb payload capacity of AAV.
  • a 530 bp enhancer sequence Dlx upstream of the promoter may be additionally included (Dimidschstein et al., 2016).
  • the cytomegalovirus CMV promoter is employed to drive mouse ATP1A3 cDNA expression.
  • the promoter sequence comprises an artificial composite promoter (P CBh ) consisting of CMV enhancer, chicken 3-actin promoter, and MVM intron (Gray et al., 2011).
  • This promoter can drive strong, albeit not cell-specific, expression in mammalian cells.
  • the promoter sequence comprises a human neuron-specific promoter (P hSyn ) with “strong, but not exclusive bias towards inhibitory neurons” (Nathanson et al., 2009). Because endogenous ATP1A3 is preferentially expressed in GABAergic (inhibitory) neurons, this promoter may be a safe choice to drive ATP1A3 expression in clinically relevant neurons of the AHC mice.
  • P hSyn human neuron-specific promoter
  • this promoter may be a safe choice to drive ATP1A3 expression in clinically relevant neurons of the AHC mice.
  • the nucleic acid regulatory sequence comprises a sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene.
  • a Kozak sequence is included to maximize the ribosome binding and translation efficiency of the supplied transgenic ATP1A3.
  • GCCACC sequence was included. This sequence represents the most common conserved RBS (ribosome binding site).
  • aCCGGAgccacc sequence (SEQ ID NO: 7) was included.
  • the sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene is included at the 3′ end of the promoters immediately in front of the ATG start codon of the subsequence ATP1A3 ORF.
  • the non-coding region 3′ to the protein-coding sequence may be retained for its transcriptional termination regulatory sequences, such as those which provide for termination and polyadenlylation.
  • the transcriptional termination signals may be provided. Where the transcriptional termination signals natively associated with the coding sequence are not satisfactorily functional in the expression host cell, then a different 3′ region, functional in the host cell, may be substituted.
  • the nucleic acid regulatory sequence comprises a 3′-UTR sequence that contains a polyadenylation sequence.
  • the 3′-UTR sequence is a rabbit beta globin polyadenylation sequence (rBGpA).
  • rBGpA rabbit beta globin polyadenylation sequence
  • rBGpA 127-bp rabbit beta globin polyadenylation sequence
  • the recombinant AAV vector comprises an AAV vector, a P ATP1A3 promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a P ATP1A3 promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • the recombinant AAV vector comprises AAV9/P ATP1A3 -ATP1A3 cDNA-rBGpA.
  • the recombinant AAV vector comprises SEQ ID NO: 3.
  • the recombinant AAV vector comprises an AAV vector, a P hSyn promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a P hSyn promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • the recombinant AAV vector comprises AAV9/P hSyn -ATP1A3 cDNA-rBGpA.
  • the recombinant AAV vector comprises SEQ ID NO: 4.
  • the recombinant AAV vector comprises an AAV vector, a P CBh promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a P CBh promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • the recombinant AAV vector comprises AAV9/P CBh -ATP1A3 cDNA-rBGpA.
  • the recombinant AAV vector comprises SEQ ID NO: 5.
  • Another aspect of the invention relates to a method for treating a subject having a neurological disorder associated with mutations in the ATP1A3 gene.
  • the method comprises administering into the central nervous system (CNS) of the subject a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 protein, thereby restoring some degree of neurological function in said subject.
  • CNS central nervous system
  • AAV adeno-associated virus
  • the neurological disorder may be selected from the group consisting of alternating hemiplegia of childhood (AHC); rapid-onset dystonia-parkinsonism (RDP); cerebellar ataxia, areflexia, pes cavus, optic atrophy, and sensorineural hearing loss (CAPOS); early infantile epileptic encephalopathy (EIEE); relapsing encephalopathy with cerebellar ataxia (RECA); dystonia, dysmorphism, encephalopathy, MRI abnormalities, no hemiplegia (D-DEMO); familial hemiplegic migraine (FHM); childhood onset schizophrenia (COS); childhood rapid onset ataxia (CROA); and auditory neuropathy spectrum disorder (ANSD). All patients with loss-of-function ATP1A3 mutations can benefit directly from the ATP1A3-based gene therapy.
  • AHC alternating hemiplegia of childhood
  • RDP rapid-onset dystonia-parkinsonism
  • CAPOS sensorineural hearing loss
  • the method can restore alpha-3-specific NaK-ATPase activity in the central nervous system, thereby restoring some degree of neurological function in symptomatic mammals (animals or humans) with mutations in ATP1A3 gene.
  • the method can restore ATP1A3 function in GABAergic neurons in the CNS, which includes Purkinje neurons of the cerebellum.
  • Another aspect of the invention relates to a method for delivering to the central nervous system of a mammal, a ATP1A3 DNA that is expressed in a cell of the central nervous system.
  • the method comprises administering into the central nervous system of the mammalian a recombinant AAV vector comprising a nucleic acid sequence encoding a ATP1A3 protein, wherein said vector transduces the cell.
  • a “subject,” “individual,” or “patient” is used interchangeably herein, which refers to a vertebrate, e.g., a mammal. Mammals include, but are not limited to, mice, rats, simians, humans, farm animals, sport animals, and pets. In certain embodiments, the subject is human.
  • the intended developmental stage (age) of the administration into the subject is neonate, pediatric, or juvenile (pre-adult).
  • This gene therapy/delivery method has been tested in various available AHC mouse models by injecting ATP1A3 cDNA-carrying AAV capsids into CSF (via intrathecal or intracisternal injection), using a similar method used to rescue adult mouse models of Mucopolysaccharidosis and diabetic neuropathy.
  • This approach provides a safer and more imminently testable approach than genomic editing, and a targeted and rapid path to long-term treatment. Because AAV does not integrate into the genome, the methods described herein also mitigate risks associated with unintended off-target effects of genomic editing.
  • the recombinant AAV vector may be administered to one or more CNS components selected from the group consisting of cerebrospinal fluid (CSF), cisterna magna (cerebellomedullaris cistern), the spinal cord, brainstem (medulla, pons, and midbrain), hippocampus, cerebellum, diencephalon (thalamus, hypothalamus), and telencephalon (corpus striatum, cerebral cortex, or, within the cortex, the occipital, temporal, parietal or frontal lobes).
  • CSF cerebrospinal fluid
  • cisterna magna Cerebellomedullaris cistern
  • the spinal cord medulla, pons, and midbrain
  • brainstem medulla, pons, and midbrain
  • hippocampus cerebellum
  • diencephalon thalamus, hypothalamus
  • telencephalon corpus striatum, cerebral
  • the recombinant AAV vector may be delivered to one or more of these CNS components via intracisternal, intrathecal, or intracerebroventricular injection.
  • the AAV vector injection can be optimized for cerebellar (Purkinje neuron) targeting.
  • Sites of AAV injection can include intravenous, intrathecal (IT), intra-cisterna magna (ICM), intraparenchymal, intraventricular, and intracerebroventricular.
  • Intravenous administration of AAV is associated with severe hepatotoxicity risk, immunological response, and is least efficient in CNS penetration due to the blood-brain barrier (Hinderer et al., 2018). Additionally, both intraventricular and intraparenchymal routes require invasive intracranial surgery, and the latter has shown very limited distribution of transduction (Hardcastle et al., 2018).
  • CSF cerebrospinal fluid
  • Administration of recombinant AAV vector directly into the cerebrospinal fluid (CSF) can deliver the vector directly to the CNS (bypassing the blood brain barrier), distribute it around and throughout the nervous system, and result in wide pattern of neuronal transduction without transfecting internal organs (Federici et al., 2012), thus bypassing immunological response and eliminating the risk of hepatic cytotoxicity associated with intravenous administration (Hinderer et al., 2018).
  • CSF delivery of AAV9 is proven safe and highly efficient in targeting the CNS in various mammals, such as adult mice and pigs.
  • the intra-cisterna magna (ICM) route of the AAV injection results in superior CNS distribution of AAV vectors compared with other routes of CSF access (Hinderer et al., 2014).
  • IT intrathecal
  • aAV9 administered directly into the CSF fluid through an intrathecal (IT) route
  • IT administration of 2 ⁇ 10 11 vg of AAV9 in pigs resulted in broad spinal motor neuron and extensive cerebellum transduction, with undetectable levels of peripheral organ transduction (Federici et al., 2011).
  • Intrathecal delivery of contrast media, anesthetics, chemotherapeutic agents and drugs for dystonia, spasticity or pain is routinely performed in human medicine.
  • even less invasive IT injection into lumbar cistern was sufficient to achieve broad and efficient transfection of the spinal cord, pons, cerebellum and other CNS parts in mice (Guo et al., 2016).
  • Intrathecal injection is desirable for AAV delivery because it is:
  • the injection dosage for the intracerebroventricular, IT, or IV administration into the CSF fluid of a mammal should be an effective dose, or effective multiple doses, of the composition comprising the recombinant AAV vector.
  • a single dose may range from 1 ⁇ 10 9 vg (vector genomes)/kg dose to 1 ⁇ 10 15 vg/kg, for instance, from 0.5 ⁇ 10 10 vg dose to 5 ⁇ 10 11 vg/kg, from 1 ⁇ 10 10 vg/kg to 5 ⁇ 10 11 vg/kg, or from 1 ⁇ 10 12 vg/kg to 1 ⁇ 10 14 vg/kg.
  • a single dose injected into a mouse brain via intracisternal or intraventricular injection
  • the total volume of injected AAV solution is, for example, between 1 to 20 ⁇ l.
  • volumes and delivery rates are appropriately scaled.
  • volumes of 150 ⁇ l can be safely injected in the primate brain (Janson et al. (2002) Hunt. Gene Ther. 13:1391-1412).
  • Treatment may consist of a single injection per target site, or may be repeated in one or more target sites. Injections can be single or multiple, unilateral or bilateral.
  • experimental results have proven that the gene-coding AAV9 vectors are capable of transducing cerebellum and increasing a protein and activity levels.
  • an endogenous mouse ATP1A3 promoter (P ATP1A3 ) was used in AAV9 vector, and the results show that expression of mCherry protein in the neurons of ventral cerebellum was detected in mice injected with the AAV9 vector with mouse P ATP1A3 promoter driving mCherry.
  • SEQ ID NO: 2 The sequence of such recombinant AAV9 is shown in SEQ ID NO: 2.
  • results have proven that the gene-coding AAV9 vectors are capable of transducing brainstem and increasing a protein levels.
  • the results show the presence of mCherry protein in mouse brainstem following injection of a recombinant vector comprising AAV9/P ATP1A3 -mCherry cDNA-rBGp(A): there were visible quantity of mCherry protein in cross-sections of the six brainstems of injected mice.
  • the sequence of such recombinant AAV9 is shown in SEQ ID NO: 2.
  • mCherry protein in mouse brainstem following the injection of AAV9/P ATP1A3 -mCherry cDNA-rBGp(A) is significant. This is because for a transgenic protein to be present in detectable amounts in the tissue, a sequential series of processes have to occur:
  • exogenous DNA construct from mouse (P ATP1A3 promoter), rabbit (rBGp(A) sequence), jellyfish (mCherry), some generic eukaryotic elements (Kozak sequence), and viral AAV2 DNA (ITR elements).
  • mouse P ATP1A3 promoter
  • rabbit rBGp(A) sequence
  • jellyfish mCherry
  • Kozak sequence some generic eukaryotic elements
  • viral AAV2 DNA ITR elements
  • An exemplary mouse promoter P ATP1A3 sequence (SEQ ID NO: 1) acatactgcaagatggtggcactctggggccctgcattactgcaattcactgggcctttcctccaccctgtatctaccccactcccagaa ggaggcagattccagggtgcctcaccctcaaagcctcggtcccctaagatacctcccctatattgagggggggtctctgagtccccaccctgg ggatgtcccgggatcaccccccccccccccccgcactgtgctcagcttctcagtggcctcagtggcttgcagaaacaaggttggagcggtgagggggg gaagggggagtacagctgcagtactgggggccgggcgcag
  • ITR2-Patpla3-mCherry-rBGp(A)-ITR2 GCCACC Kozak sequence Cctgcaggcagctgcgcgctcgctcactgaggccgcccgggcaaagcccgggcgacctttggtcgcccggcctcagt gagcgagcgagcgcgcagagagggagtggccaactccatcactaggggttcct gcggccgcaCGCGT acatactgcaagatggtggcactctggggccctgcattactgcaattcactgggcctttcccccaccctgtatctaccccactcccagaa ggaggcagattccagggtgcctcaccctcaaagcctccctaagatacctcccctatat
  • the promoter sequence is indicated in lower case letters and marked in boldface. This part of the cassette drives transcription of the transgene and defines the cellular specificity of its expression.
  • mouse promoter Patp1a3 was used, and may be replaced with the corresponding homologous human sequence for human application, the mouse promoter.
  • the ATP1A3 protein-coding sequence is the same in all three sequences, indicated in capital letters, and is represented by the open reading frame (ORF) of the most predominant transcript of mouse ATP1A3 gene.
  • this mouse ATP1A3 protein-coding sequence may be replaced with ORF of the corresponding human sequence, which is also the predominant transcript of human ATP1A3 gene.
  • This human ORF sequence is below (SEQ ID NO: 6).
  • mice models of AHC are available for gene therapy testing in vivo.
  • Six distinct mouse strains carrying common AHC-associated ATP1A3 alleles have been generated and characterized. These mouse models phenocopy many clinical symptoms of AHC, demonstrating causality and reproducing the disease severity spectrum observed in AHC patients. These strains thus provide multiple, available, and independent experimental substrates to test the effectiveness of proposed gene delivery in vivo.
  • D801N/+ mice were generated via CRISPR-based genomic editing. When introduced into mice, the D801N allele (GAC ⁇ AAC) also contains the PAM-blocking silent L8202L mutation (CTG ⁇ CTA).
  • I810N/+ frozen embryos were provided by the strain creator, Dr. Steven Clapcote.
  • mouse primary cerebellar (Purkinje cell) culture is transfected with AAV vector carrying 500 pb mouse ATP1A3 promoter and 3 kb cDNA.
  • Controls include un-transfected neuron culture, mock transfected neuron culture, and neuron culture transfected with AAV with ATP1A3 promoter-driven GFP.
  • Example 1A Determining Whether Delivery of Wild Type ATP1A3 Gene Via CSF is Sufficient to Rescue Mouse ATP1A3 KO Model of AHC
  • KO AHC mice models (DeAndrade, or Ikeda a3+/AE2-6 mice, or Lingrel a3+/KOI4 mice) with ATP1A3 carrying AAV vector. Neonates, 2 week old, and 2 month old mice are tested.
  • Example 1B Determining Whether Delivery of Wild Type ATP1A3 Gene to CSF is Sufficient to Rescue Mouse ATP1A3 Missense Model of AHC
  • Inject available missense AHC mouse models (D801N, D801Y, I810N and E815K) at site determined in (1) above with ATP1A3 carrying AAV vector. Neonates, 2 week old, and 2 month old mice are tested. The test for rescue is as described for KO mice above.
  • This example compares specificity and strength of ATP1A3 transgene expression driven by three different AAV9 vectors in mouse brains (AAV vector constructs differ only in their promoter configuration, all the rest of the coding and regulatory DNA sequences are identical among them).
  • Vector biodistribution and expression specificity was determined by double labeling with antibodies against mouse ATP1A3 protein with specific neuronal markers such as Calbindin for Purkinje neurons and GAD67 for GABA-ergic neurons.
  • the first steps consisted of determining the best antibodies and staining conditions to detect ATP1A3 in mouse brain tissues, and in neuronal subtypes: Purkinje cells in the cerebellum and GABAergic neurons in cortex, hippocampus and basal ganglia.
  • mice injected with mCherry and ATP1A3 expressing AAV viruses, as shown in Table 1, below.
  • AAV9_PCBh :ATP1A3 Nov. 29, 2018 4-6 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 29, 2018 4-7 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 30, 2018 4-8 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 30, 2018 1.
  • Brains received by Novaxia were pre-sliced to isolate the cerebellum, which was dehydrated and included in paraffin blocks. 4 ⁇ m slices were obtained from each cerebellum and put on glass slides.
  • ATP1A3 antibodies were tested on hippocampal and cerebellum slices.
  • the calbindin antibody was tested on cerebellum slices and the GAD67 antibody was tested on hippocampal slices.
  • ATP1A3 a secondary anti-rabbit Alexa 488 antibody was used for detection while for Calbindin and GAD67 and anti-chicken Alexa 594 antibody was used (Invitrogen, 1:400 dilution).
  • mCherry detection as mCherry is a red fluorescent protein, an Alexa 594 anti-rat antibody was chosen for mCherry (1:400 dilution).
  • mouse cerebella which were injected with AAV viruses
  • antigen retrieval was performed at pH8 for all antibodies, and slides were labeled with the anti-mCherry antibody (Molecular Probes #M11217) at 1:200+anti-Calbindin antibody (Synaptic Systems #214006) at 1:200, and anti-ATP1A3 antibody (EMD Millipore antibody #06-172-1) at 1:250+anti-Calbindin antibody (Synaptic Systems #214006) at 1:200.
  • Staining involved the following categories of antibodies: ATP1A3 antibodies, Calbindin antibodies, GAD67 antibodies, and mCherry antibodies.
  • FIG. 1 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum. This ATP1A3 elicitates a good specific signal compared to the signal obtained in the absence of primary antibody, at both antigen retrieval pH. However, a higher signal was obtained with antigen retrieval at pH8. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 2 depicts ATP1A3 (Millipore #06-172-1) staining in mouse cerebellum.
  • the zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition, showing the widespread labeling and higher labeling fluorescence in what appears to be Purkinje cells.
  • FIG. 3 depicts ATP1A3 (Millipore #06-172-1) staining in mouse brain at dorsal hippocampus level.
  • This ATP1A3 antibody elicitates a good specific signal compared to the signal obtained in the absence of primary antibody, at both antigen retrieval pH. However, a higher signal was obtained with antigen retrieval at pH8. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 4 depicts ATP1A3 (Millipore #06-172-1) staining in mouse brain at dorsal hippocampus level.
  • the zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition, showing the widespread labeling.
  • Some cells appear to have higher perinuclear signals in the parietal cortex and in the hippocampus in CA1, CA2-3 and Dentate Gyrus.
  • FIG. 5 shows the ATP1A3 (Millipore #06-172-1) staining in the dorsal hippocampus.
  • the zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition (pH8 antigen retrieval) in the dorsal hippocampus. Arrows show cells with higher labeling.
  • FIG. 6 shows the ATP1A3 (Millipore #06-172-1) staining in the parietal cortex.
  • the zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition (pH8 antigen retrieval), in the parietal cortex. Arrows show examples of cells with higher labeling.
  • FIG. 7 depicts Calbindin (Synaptic Systems #214006) staining in mouse cerebellum. This antibody elicited the best specific signal over the negative control in the pH8 antigen retrieval condition.
  • FIG. 8 depicts Calbindin (Synaptic Systems #214006) staining in mouse cerebellum.
  • the zoomed image shows Purkinje cells labeled by the calbindin antibody using the pH8 antigen retrieval condition.
  • FIG. 9 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level.
  • FIG. 10 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol.
  • the zoomed image over the dorsal hippocampus shows the detection of GABAergic neurons with low fluorescence levels.
  • FIG. 11 depicts GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level.
  • FIG. 12 depicts GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol.
  • the zoomed image over the dorsal hippocampus (pH8 antigen retrieval protocol) shows good detection of GABAergic neurons in the hippocampus and in the cortex.
  • FIG. 13 depicts mCherry staining in mouse cerebellum. Intense stainings are seen below the fourth ventricle and around this region. In the cerebellum, the staining is more diffuse, but specific staining is observed compared to negative brains (infected with ATP1A3 AAV viruses).
  • FIG. 14 depicts the mouse cerebellum anatomy.
  • VERM indicating the Vermal regions, includes: LING Lingula (I); CENT Central lobule; CUL Culmen; DEC Declive (VI); FOTU Folium-tuber vermis (VII); PYR Pyramus (VIII); UVU Uvula (IX); and NOD Nodulus (X).
  • HEM indicating the Hemispheric regions, includes: SIM Simple lobule; AN Ansiform lobule; PRM Paramedian lobule; COPY Copula pyramidis; PFL Paraflocculus; and FL Flocculus.
  • CBN indicating Cerebellar nuclei, includes: FN Fastigial nucleus; IP Interposed nucleus; DN Dentate nucleus; and VeCB Vestibulocerebellar nucleus.
  • FIG. 15 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition.
  • the zoomed images are of the red channel. Intense stainings are seen below the fourth ventricle and around this region. In the cerebellum, the staining is more diffuse with occasionally a more intense cell that could be a Purkinje cell.
  • FIG. 16 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition.
  • the zoomed images are of the red channel at levels of Purkinje cell layers. Arrows show examples of Purkinje cells.
  • the Millipore #06-172-1 antibody demonstrated that it was able to detect the target protein, detecting a similar signal as disclosed in the Bottger et al. (2011) publication, in which the specificity of this antibody was shown.
  • Calbindin was well detecting by the tested antibody, giving signals that can be segmented for Purkinje cell detection. The best condition was to use antigen retrieval at pH8.
  • the GAD67 antibody from Synaptic Systems (#198 006) allowed detecting some GABAergic neurons but with low fluorescence levels over background.
  • the GAD67 antibody from Millipore (#MAB5406) allowed detecting GABAergic neurons with good sensitivity and fluorescence levels and can thus be used for further steps of the study plan.
  • the mCherry antibody allowed detecting positive cells only in the mCherry infected group, with strong signals below the fourth ventricle but also labelings in other regions. Co-labeling with the Calbindin antibody will allow determining if specific signals are measured in Purkinje cells. The signals in Purkinje cells are not as strong as below the fourth ventricle.
  • Calbindin signals allowed determining the localization of Purkinje neurons.
  • the ATP1A3 signals were peripheral to cells, so once cells were segmented, ATP1A3 was measured in a ring around the calbindin signal.
  • FIG. 17 Cell-by-cell data was fed into the proprietary Cytosurfer software to make cytometric analyses.
  • the mean ATP1A3 fluorescence was plotted against cell count ( FIG. 17 ) and the cells from the same groups displayed group by group. The intensity histograms were well superposed.
  • a gate was chosen to detect Purkinje cells with higher ATP1A3 intensity ( FIG. 18 ) which should appear if ATP13 intensity is increased.
  • FIGS. 19 , 20 , and 21 show the detected Purkinje neurons and the localization of cells with higher mean intensity. These cells are anatomically regularly spread and do not present a gradient from the cisterna magna.
  • FIG. 21 most of the cells detected with increased ATP1A3 are false positive due to creases in the cerebellum slice which induces a higher green fluorescence signal.
  • mCherry expression was used as a positive control of AAV injection and transgene expression. Cytometric analysis of mCherry intensity in Purkinje neurons showed a similar profile in mCherry infected mice compared to ATP1A3 infected mice ( FIG. 24 ). A few cells with higher mCherry fluorescence were detected, but this was artificial as these higher signals appeared where creases in the tissue were present ( FIG. 25 ). Quantifications support the absence of differences between treatment groups in Purkinje Neurons ( FIG. 26 ) and other cells ( FIG. 27 ).
  • mCherry staining was well detected in some brainstems of mCherry-AAV infected brains while no specific signals were present in ATP1A3-AAV infected brains.
  • the pattern of mCherry expression was not widespread but instead focalized in discrete regions, which suggests that the injection site was not truly in the Cisterna Magna but rather in the brainstem itself.
  • no mCherry expression could be detected ( FIG. 28 ).
  • AAV9_PAT ATP1A3 P1A3 infected mCherry brains 127 98.6 123 132 93.9 106 164 118 113 90.2 93.3 86.9 149 123
  • ATP1A3 mean fluorescence in subpopulation with higher ATP1A3 AAV9_PAT AAV9_PAT AAV9_ AAV9 P1A3:: P1A3:: Psynapsin:: PCBh:: mCherry ATP1A3 ATP1A3 ATP1A3 0 12 12 6.579797 5.997402 5.9995585 7.1953383 8.75 5.848485 0 6.5679874 6.0148131 8.8166668 7.1851386 6.0072859 7.1872419 10 7.1648367 7.183503 7.1800464 0 0 6.0020599 6.1937063 0 8.5209115 7.1846988 6.5676226 0 0 7.177844 6.0161244

Abstract

This invention relates to a transgenic vector for transducing cells of a mammal's CNS and transgenically expressing a protein in the mammal's CNS. The transgenic vector comprises a virus-derived vector, a nucleic acid sequence encoding the protein, and an endogenous ATP1A3 promoter sequence. This invention also relates to a composition comprising a recombinant AAV vector comprising a nucleic acid sequence encoding a ATP1A3 protein, in a form compatible with administration into the CNS. The invention also relates to a method for treating a subject having a neurological disorder associated with mutations in the ATP1A3 gene and a method for delivering a transgenic ATP1A3 DNA to the central nervous system of a mammal by administering the recombinant AAV vector into the mammal's CNS.

Description

  • This application is a continuation application of U.S. patent application Ser. No. 16/379,440, filed Apr. 9, 2019, which claims priority, under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 62/654,645, filed on Apr. 9, 2018, which is herein incorporated by reference in its entirety.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been filed electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Aug. 10, 2023, is named 24892_0200-US-CON_SL.xml and is 25,770 bytes in size.
  • FIELD OF THE INVENTION
  • The present invention relates to the delivery and expression of ATP1A3 genes in the central nervous system.
  • BACKGROUND OF THE INVENTION
  • Alternating Hemiplegia of Childhood (AHC) is a serious and rare neurodevelopmental disorder, affecting approximately 1 in 1,000,000 children. AHC is caused by de novo loss-of-function mutations in the ATP1A3 gene (Heinzen et al., 2012; Rosewich et al., 2012). The gene encodes the α3 subunit of the Na+, K+ ATPase, one of two pumps that establish and maintain electrochemical gradients required for the propagation of action potentials in neurons. AHC-associated mutations do not affect expression of ATP1A3 gene or levels of its protein, but significantly reduce α3 protein functionality (Heinzen et al., 2012; Li et al., 2015). Two most common AHC-associated alleles of ATP1A3 cause D801N and E815K missense substitutions (Heinzen et al., 2012; Rosewich et al., 2012; Sasaki et al., 2014; Viollet et al., 2015).
  • ATP1A3 gene is expressed in the central nervous system (CNS) and heart of all mammals studied. Brain expression of ATP1A3 is enriched in high frequency GABAergic interneurons of the cerebellum, pons, basal ganglia, thalamus, cortex and hippocampus (Bøttger et al., 2011; Hieber et al., 1991; McGrail et al., 1991; Pietrini et al., 1992; Richards et al., 2007). While most neurons of the CNS also express ATP1A1 gene that can partially compensate for the loss of ATP1A3 functionality, some neuron types, most notably Purkinje cells of the cerebellum and certain inhibitory interneurons with high-frequency firing rate, do not express ATP1A1, leaving them especially vulnerable to ATP1A3 dysfunction. Clinical symptoms of cerebellar, hippocampal, basal ganglia, and motor cortex dysfunction largely correlate with persistent symptoms of AHC. In line with clinical symptoms, brains of AHC patients show progressive frontal dominant cerebral, severe hippocampal, and diffuse cerebellar atrophy (Sasaki et al., 2017). Paroxysmal symptoms of AHC likely originate in Purkinje cells and spread through the afferent networks of the cerebellum by spreading depolarization (“spreading depression”).
  • AHC symptoms appear before 18 months of age, and often shortly after birth. AHC causes severe morbidity and increased mortality. Patients often suffer severe neurodevelopmental impairments, episodes of paralysis, severe dystonia, ataxia, swallowing problems, epilepsy, status epilepticus, sudden unexpected death, and episodes of developmental regression (Mikati et al., 2000; Panagiotakaki et al., 2015). Currently, there is no effective on-target treatment, although certain off-target drugs like Flunarizine have been reported to have some limited effect on certain symptoms.
  • Besides AHC, mutations in ATP1A3 gene are associated with at least eight other distinct, but clinically overlapping neurological disorders (Dard et al., 2015; de Carvalho Aguiar et al., 2004; Demos et al., 2014; Heinzen et al., 2014; Paciorkowski et al., 2015; Smedemark-Margulies et al., 2016; Sweadner et al., 2016; Sweney et al., 2015; Yano et al., 2017). The clinical symptoms of these diseases partly intersect and range from mild to severe. The number of patients with pathogenic ATP1A3 alleles is likely to increase if ATP1A3 is included in gene panels for epilepsy-like disorders, as these patients are often misdiagnosed (Algahtani et al., 2017).
  • There thus remains a need in the art to develop an on-target delivery of ATP1A3 genes and an effective gene therapy treatment of neurological disorders associated with loss-of-function mutations in ATP1A3. This invention answers this need.
  • SUMMARY OF THE INVENTION
  • One aspect of the invention relates to a transgenic vector for transducing cells of the central nervous system of a mammal and transgenically expressing a protein in the central nervous system of the mammal. The transgenic vector comprises a virus-derived vector, a nucleic acid sequence encoding the protein, and an endogenous ATP1A3 promoter (PATP1A3) sequence.
  • One aspect of the invention relates to a composition comprising a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3) protein, in a form compatible with administration into the central nervous system.
  • Another aspect of the invention relates to a method for treating a subject having a neurological disorder associated with mutations in the ATP1A3 gene. The method comprises administering into the central nervous system (CNS) of the subject a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 protein, thereby restoring some degree of neurological function in said subject.
  • Another aspect of the invention relates to a method for delivering to the central nervous system of a mammal, a ATP1A3 DNA that is expressed in a cell of the central nervous system. The method comprises administering into the central nervous system of the mammalian a recombinant AAV vector comprising a nucleic acid sequence encoding a ATP1A3 protein, wherein said vector transduces the cell.
  • Additional aspects, advantages and features of the invention are set forth in this specification, and in part will become apparent to those skilled in the art on examination of the following, or may be learned by practice of the invention. The inventions disclosed in this application are not limited to any particular set of or combination of aspects, advantages and features. It is contemplated that various combinations of the stated aspects, advantages and features make up the inventions disclosed in this application.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 2 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum, as a zoomed image.
  • FIG. 3 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse brain at dorsal hippocampus level. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 4 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse brain at dorsal hippocampus level, as a zoomed image.
  • FIG. 5 depicts ATP1A3 (Millipore #06-172-1) staining in the dorsal hippocampus, as zoomed image.
  • FIG. 6 depicts ATP1A3 (Millipore #06-172-1) staining in the parietal cortex, as a zoomed image.
  • FIG. 7 depicts Calbindin (Synaptic Systems #214006) staining in a mouse cerebellum.
  • FIG. 8 depicts Calbindin (Synaptic Systems #214006) staining in a mouse cerebellum, as a zoomed image.
  • FIG. 9 depicts GAD67 (Synaptic Systems #198 006) staining in a mouse brain at dorsal hippocampus level.
  • FIG. 10 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol, as a zoomed image over the dorsal hippocampus.
  • FIG. 11 depicts GAD67 (Millipore #MAB5406) staining in a mouse brain at dorsal hippocampus level.
  • FIG. 12 depicts: GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol.
  • Zoomed image over the dorsal hippocampus (pH8 antigen retrieval protocol) showing good detection of GABAergic neurons in the hippocampus and in the cortex.
  • FIG. 13 depicts mCherry staining in a mouse cerebellum.
  • FIG. 14 depicts the mouse cerebellum anatomy.
  • FIG. 15 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition, as zoomed images.
  • FIG. 16 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition, as zoomed images.
  • FIG. 17 depicts histograms of the mean intensity of ATP1A3 in the 4 experimental groups. Histograms are well superposed.
  • FIG. 18 depicts histograms of the mean intensity of ATP1A3 in the 4 experimental groups with the gate for highest fluorescence levels.
  • FIG. 19 depicts an example of Purkinje cell detection. Contours of detected cells are shown.
  • FIG. 20 depicts an example of Purkinje cell detection showing cells with higher ATP1A3 levels. Contours of detected cells are shown in dark shading, and cells in gate are shown in light shading.
  • FIG. 21 depicts an example of Purkinje cell detection showing cells with higher ATP1A3 levels. Contours of detected cells are shown in dark shading, and cells in gate are shown in light shading.
  • FIG. 22 depicts graphical representations of mean ATP1A3 intensity in Purkinje Neurons and percentage of Purkinje neurons with high ATP1A3 intensity. The left graft shows mean intensity in Purkinje Neurons, and the right graph shows the percentage of Purkinje neurons with High ATP1A3 intensity.
  • FIG. 23 depicts graphical representations of mean ATP1A3 intensity in cerebellar cells other than Purkinje neurons.
  • FIG. 24 depicts a histogram of the mean intensity of mCherry in the mCherry group and animals from ATP1A3 infected groups, with gate for highest fluorescence levels.
  • FIG. 25 depicts an example of Purkinje cell detection showing cells with higher mCherry levels (cells in Gate named Highest). Contours of detected cells are shown in black, and cells in gate are shown in dark shading.
  • FIG. 26 depicts graphical representations of mean mCherry intensity in Purkinje Neurons and the percentage of Purkinje neurons with high mCherry intensity. The left graft shows the mean intensity in Purkinje Neurons. The right graph shows the percentage of Purkinje neurons with High mCherry intensity.
  • FIG. 27 depicts graphical representations of mean mCherry intensity in cerebellar cells other than Purkinje neuron.
  • FIG. 28 depicts counts of mCherry positive cells in mCherry infected brainstems.
  • FIG. 29 depicts an example of cells with higher ATP1A3.
  • FIG. 30 depicts quantification of ATP1A3 immunofluorescence. The left graft shows ATP1A3 immunofluorescence at the surrounding of all detected cells. The middle graft shows the percentage of cells detected as higher expressing ATP1A3 after gating. The right graft shows mean fluorescence in cells in highest ATP1A3 gate.
  • DETAILED DESCRIPTION OF THE INVENTION
  • One aspect of the invention relates to a transgenic vector for transducing cells of the central nervous system of a mammal and transgenically expressing a protein in the central nervous system of the mammal. The transgenic vector comprises a virus-derived vector, a nucleic acid sequence encoding the protein, and an endogenous ATP1A3 promoter (PATP1A3) sequence.
  • Suitable viral vectors include those neurotrophic viral vectors, including, but not limited to, adeno-associated viral vectors (AAV), herpes simplex viral vectors and lentiviral vectors. More discussions about the herpes simplex viral vectors may be found in U.S. Pat. No. 5,672,344, which is incorporated herein by reference in its entirety.
  • All the descriptions and embodiments relating to the AAV vectors discussed in a different aspect of the invention, infra, are suitable for this aspect of the invention.
  • The transgenic vector also comprises a nucleic acid sequence encoding a protein (DNA encoding the protein, e.g., cDNA of protein), the transgene. The transgene encodes a biologically active protein which is transgenically expressed in the central nervous system of a mammal. The transgene may be ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3). The transgenes may also be insulin growth factor-1 (IGF-1), calbindin D28, parvalbumin, HIF1-alpha, SIRT-2, VEGF, SMN-1, SMN-2, CNTF (Ciliary neurotrophic factor), sonic hedgehog (shh), erythropoietin (EPO), lysyl oxidase (LOX), progranulin, prolactin, ghrelin, neuroserpin, or placenta lactogen.
  • The protein-coding nucleic acid may be an DNA sequence, an “exogenous DNA” that is exogenous to both AAV and to the target cell. The DNA may be synthetic DNA, complementary DNA, genomic DNA, or a combination thereof. The DNA may be of any sequence or length, provided that it may be incorporated into the vector and delivered to target cells. Typically, because of the packaging limitations of AAV, the exogenous DNA will have a length of up to 4,400 bases.
  • More than one transgene can be delivered by the transgenic vector, e.g., via more than one virus-derived vector. For instance, the transgenic vector comprises multiple AAV vectors and each AAV vector comprises a transgene operably linked to a promoter.
  • The protein-coding nucleic acid is operably linked to a promoter sequence to enable the expression of the trangene from a single AAV vector. For the gene to be expressible, the protein-coding nucleic acid shall be operably linked to a promoter sequence so that when the promoter is activated, the coding sequence is transcribed. The protein-coding nucleic acid is operably linked if the linkage does not cause an error in the reading of the downstream sequence. To be “operably linked,” it is not necessary that two sequences be immediately adjacent to one another.
  • The promoter sequence drives transcription of the transgene and defines the cellular specificity of its expression. The promoter sequence is an endogenous ATP1A3 promoter (PATP1A3) sequence. In one embodiment, the PATP1A3 promoter comprises a mouse promoter sequence, SEQ ID NO: 1. In one embodiment, the PATP1A3 promoter comprises a homologous human sequence.
  • The transgenic vector may further comprise one or more nucleic acid regulatory sequence, linked directly or indirectly to the protein-coding nucleic acid sequence. For instance, a promoter specific to the gene may be incorporated into the transgenic vector. In certain embodiments, the nucleic acid regulatory sequence comprises a sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene. In certain embodiments, the nucleic acid regulatory sequence comprises a 3′-UTR sequence that contains a polyadenylation sequence.
  • All the descriptions and embodiments relating to the nucleic acid regulatory sequences discussed in a different aspect of the invention, infra, are suitable for this aspect of the invention.
  • In certain embodiments, the transgenic vector comprises an AAV vector (e.g., a recombinant AAV vector), a PATP1A3 promoter, and a mCherry transgene.
  • In certain embodiments, the transgenic vector comprises AAV9 vector (e.g., recombinant AAV9 vector), a PATP1A3 promoter, a mCherry transgene, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence. In one embodiment, the transgenic vector comprises SEQ ID NO: 2.
  • In certain embodiments, the transgenic vector comprises an AAV vector (e.g., a recombinant AAV vector), a PATP1A3 promoter, and a ATP1A3 transgene.
  • In certain embodiments, the transgenic vector comprises AAV9 vector (e.g., recombinant AAV9 vector), a PATP1A3 promoter, a ATP1A3 transgene, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence. In one embodiment, the transgenic vector comprises SEQ ID NO: 3.
  • Also disclosed herein are compositions and methods for intracisternal or intrathecal administration of recombinant AAV comprising a gene that affects neurological function. The embodiments of the invention illustrate that some neurological disorders associated with loss-of-function mutations of ATP1A3 genes (e.g., alternating hemiplegia of childhood, “AHC”) can be better addressed with a more targeted delivery of the gene therapy. For instance, the phenotype of an AHC mouse model has been rescued by transgenesis. Inclusion of extra-chromosomal copies of ATP1A3 by transgenesis has been shown to be sufficient to increase Na+, K+ ATPase activity in the brain and confer significant phenotypic improvements compared to non-transgenic mice with the same AHC associated mutation. This demonstrates that the presence of additional copies of functional ATP1A3 genes can compensate for lack of function in a disease-associated genomic allele of ATP1A3 and rescue the associated phenotypes.
  • One aspect of the invention relates to a composition comprising a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 (sodium/potassium-transporting ATPase subunit alpha-3) protein, in a form compatible with administration into the central nervous system.
  • AAV Vector
  • The composition contains an adeno-associated virus (AAV) vector. The vector may be a derivative of the adeno-associated virus, into which exogenous DNA is introduced.
  • One purpose is to rescue the phenotypes caused by ATP1A3 insufficiency by delivering additional copies of functional ATP1A3 cDNA to the neurons of the CNS. AAV vector is used herein as a carrier to deliver ATP1A3 cDNA.
  • AAV is a replication-defective, non-enveloped parvovirus, present in humans and some other primate species but not associated with any pathology. Recombinant AAV vectors are used for the development of human therapeutics because they can infect quiescent non-dividing cells (like neurons), mediate long-term gene expression, and, unlike retroviruses, persist in an extrachromosomal state without integrating into the genome of the host cell. Since AAV does not integrate into the host genome, the use of this viral vector mitigates risks associated with unintended off-target effects of genomic editing (Schaefer et al., 2017; Zhang et al., 2015).
  • AAV vectors are derived from single-stranded (ss) DNA parvoviruses that are nonpathogenic for mammals. Briefly, recombinant AAV-based vectors have two open reading frames, the rep and cap viral genes that account for 96% of the viral genome removed, leaving the two flanking 145-basepair (bp) inverted terminal repeats (ITRs), which are used to initiate viral DNA replication, packaging and integration. These ITRs allow for synthesis of the complementary DNA strand. Rep and Cap are translated to produce multiple distinct proteins. When constructing an AAV transfer plasmid, the transgene is placed between the two ITRs, and Rep and Cap are supplied in trans. In the absence of helper virus, wild-type AAV is typically maintained episomally. A single AAV particle can accommodate up to 5 kb of ssDNA, therefore leaving about 4.5 kb for a transgene and regulatory elements, which is typically sufficient. However, trans-splicing systems as described, for example, in U.S. Pat. No. 6,544,785, which is incorporated herein by reference in its entirety, may nearly double this limit.
  • In one embodiment, small (4.8 kb) ssDNA AAV genome having ITR sequences flanking the insert are of ITR2 serotype.
  • AAV vector of any serotype can be used. Particular useful serotypes are those that are particularly suitable for nervous cells, e.g., AAV1, AAV2, AAV4, AAV5, AAV8, AAV9, AAV-PHP.B, or AAVrh10. Several of these AAV serotypes have been utilized in clinical Phase I and II studies for diseases that require CNS delivery (reviewed in (Naso et al., 2017)). Initial testing of AAV2-based RPE65 delivery has been done in canine culture. Other serotype besides those listed herein can be used. Furthermore, pseudotyped AAV vectors may also be used herein. Pseudotyped AAV vectors are those which contain the genome of one AAV serotype in the capsid of a second AAV serotype; for example, an AAV vector that contains the AAV2 capsid and the AAV1 genome or an AAV vector that contains the AAV5 capsid and the AAV 2 genome.
  • Those skilled in the art will be familiar with the preparation of functional AAV-based gene therapy vectors. Methods of AAV production, purification, and preparation for administration to human subjects can also be found in the art (see, e.g., Viral Vectors for Gene Therapy: Methods and Protocols, ed. Machida, Humana Press, 2003, which is incorporated herein by reference in its entirety).
  • Additionally, AAV-based gene therapy targeted to cells of the CNS has been described in U.S. Pat. Nos. 6,180,613 and 6,503,888, which are incorporated by reference in their entirety.
  • Exemplary AAV vectors are recombinant AAV vectors encoding human protein.
  • In certain embodiments, the AAV vector is AAV9 vector. In these embodiments, AAV9 vector is used to deliver a rescue gene to the CNS neurons (e.g., in a mammal, such as mouse, primate, human). Cell-specificity of AAV infection is determined by the capsid serotype. AAV serotype 9 (AAV9) has natural tropism for neurons and was successfully applied to rescue adult mouse models of diseases such as mucopolysaccharidosis (Fu et al., 2010; Hinderer et al., 2016; Watson et al., 2006) and diabetic neuropathy (Homs et al., 2014). Injection of AAV9 into cerebrospinal fluid has been highly efficient in targeting the CNS in adult mice (Bey et al., 2017; Fu et al., 2010) and pigs (Federici et al., 2012). Large comparative analysis of AAV serotypes 1, 2, 5, 8 and 9 in mouse and macaque brains showed that AAV9 exhibited high efficiency cortical neuron transduction (>70%) when used in conjunction with neuron-specific promoter (Watakabe et al., 2015). AAV9 vectors showed preferentially higher selectivity of neuronal transduction in adult non-human primate compared to other serotypes (El-Shamayleh et al., 2017).
  • In an illustrative embodiment, the gene therapy is conducted in various mouse models (that phenocopy many clinical symptoms of AHC) by injecting ATP1A3 DNA-carrying AAV9 capsids into cerebrospinal fluid (via injection into cisterna magna), using a similar method used to rescue adult mouse models of Mucopolysaccharidosis (Hinderer et al., 2016; Watson et al., 2006; DiRosario et al., 2010) and diabetic neuropathy (J. Homs et al., 2014).
  • ATP1A3 Coding Sequence
  • The ATP1A3-coding nucleic acid may be a DNA sequence, an “exogenous DNA” that is exogenous to both AAV and to the target cell. For gene therapy of any genetically-based or genetically-acquired neurological disorder in a subject, DNA delivered to that subject may be considered exogenous even though it is identical to a gene native to that subject's species, provided it differs in the regulatory or coding region from the cognate gene of the subject to whom it is delivered, and therefore encodes a different gene product or is expressed to a different degree and/or in different cells, under at least some conditions.
  • The DNA may be synthetic DNA, complementary DNA, genomic DNA, or a combination thereof. The DNA may be of any sequence or length, provided that it may be incorporated into the vector and delivered to target cells. Typically, because of the packaging limitations of AAV, the exogenous DNA will have a length of about 10-5,000 bases, e.g., 100 to 4,000 bases. The size of a typical ATP1A3 cDNA (˜3 kb) fits within the carrying capacity of AAV vectors (4.4 kb).
  • In one embodiment, a mouse ATP1A3 cDNA sequence is used in ATP1A3-AAV9 vector for gene therapy used in mouse for compatibility. For instance, a sequence of the most common transcript isoform of mouse ATP1A3 gene, NCBI reference sequence NM_001290469.1, may be used. In this embodiment, only the ORF portion of the mRNA is included: from ATG start to TGA stop codons (including the latter).
  • In one embodiment, a human ATP1A3 cDNA sequence is used in ATP1A3-AAV9 vector for gene therapy used in human for compatibility. For instance, a sequence of the most abundant transcript of human ATP1A3 gene, NCBI reference sequence NM_152296.4 (SEQ ID NO: 6), may be used.
  • In one embodiment, no tags are added to its coding sequence in order not to interfere with transgenic ATP1A3 protein translation, folding, transport and function.
  • Regulatory Sequences
  • In the CNS gene therapy applications, it is necessary to control transcriptional activity. Pharmacological regulation of gene expression with the viral vectors can been obtained by including various regulatory elements as described below. The precise nature of regulatory regions needed for gene expression may vary from organism to organism.
  • In certain embodiments, the composition further comprises one or more nucleic acid regulatory sequences, linked directly or indirectly to the ATP1A3-coding nucleic acid sequence.
  • Promoter
  • In certain embodiments, the nucleic acid regulatory sequence comprises a promoter sequence that renders the expression of the ATP1A3-coding nucleic acid sequence CNS-specific. The promoters have the ability to drive the ATP1A3 expression in target areas in central nervous system. The promoter sequence can be neuron-specific or glia-specific.
  • In some embodiments, a promoter specific to the gene is incorporated into the AAV construct. Inclusion of gene-specific promoters in recombinant AAVs for gene delivery provides the desired specificity for neuronal transduction and expression within particular target cells in the brain that is not possible with promoters with non-specific expression profile, like CMV (Watakabe et al., 2015). The use of specific promoters to restrict transgene expression was utilized successfully in several gene delivery studies on non-human primates. A 300 bp TH promoter in AAV vector was used to drive expression in dopaminergic neurons of macaques, achieving a specificity of 95% (Stauffer et al., 2016). Similarly, short 530 bp enhancer sequence Dlx was used to drive GFP expression in GABAergic neurons of marmosets, achieving a specificity of 93% (Dimidschstein et al., 2016). lkb-long L7/Pcp2 promoter was used to drive ChR2 expression in Purkinje neurons with 96% selectivity (El-Shamayleh et al., 2017).
  • In certain embodiments, the promoter sequence comprises an endogenous ATP1A3 promoter (PATP1A3); a human neuron-specific promoter with a strong bias towards inhibitory neuron (PhSyn); or an artificial composite promoter comprising CMV enhancer, chicken 3-actin promoter, and MVM intron (PCBh).
  • In some embodiments, an endogenous ATP1A3 promoter is employed. In one embodiment, a pig ATP1A3 promoter is employed. In one embodiment, a mouse ATP1A3 promoter is employed (see, e.g., SEQ ID NO: 1). In one embodiment, a primate ATP1A3 promoter is employed. In one embodiment, a human ATP1A3 promoter is employed.
  • In an illustrative embodiment, the promoter sequence comprises an endogenous ATP1A3 promoter (PATP1A3) consisting of −407/+143 of mouse genomic sequence where “/” indicates the transcription start site. Mouse ATP1A3 −407/+143 promoter sequence is based on minimal pig and rat ATP1A3 promoters that were sufficient for neuronal expression in vivo (Benfante et al., 2005; Henriksen et al., 2013; Pathak et al., 1994). To avoid redundancy in having two ribosome binding sites in the constructs, the promoter is shortened to +143 to exclude endogenous Kozak sequence immediately adjacent to the start codon. This minor shortening may not have a significant negative effect on transgene expression, because −210/+77 rat sequence of ATP1A3 promoter was sufficient to drive brain-specific transgene expression in mice in vivo (Pathak et al., 1994).
  • In another illustrative embodiment, the promoter sequence comprises 488 bp-long promoter of porcine ATP1A3 closely homologous to human, which was sufficient to drive GFP expression in Zebrafish brain and spinal cord (Henriksen et al., 2013). Because porcine ATP1A3 promoter was sufficient to drive expression in evolutionarily-distant Zebrafish, it may be sufficient to drive expression on closer-related mammal, like the mouse. In total, 550 nt promoter+3 kb cDNA+133nt rabbit beta globin polyA sequence totals 3,683nt and thus fits within the 4.4 kb payload capacity of AAV.
  • Alternatively, in some embodiments, a 530 bp enhancer sequence Dlx upstream of the promoter may be additionally included (Dimidschstein et al., 2016).
  • In other embodiments, the cytomegalovirus CMV promoter is employed to drive mouse ATP1A3 cDNA expression.
  • In another illustrative embodiment, the promoter sequence comprises an artificial composite promoter (PCBh) consisting of CMV enhancer, chicken 3-actin promoter, and MVM intron (Gray et al., 2011). This promoter can drive strong, albeit not cell-specific, expression in mammalian cells.
  • In another illustrative embodiment, the promoter sequence comprises a human neuron-specific promoter (PhSyn) with “strong, but not exclusive bias towards inhibitory neurons” (Nathanson et al., 2009). Because endogenous ATP1A3 is preferentially expressed in GABAergic (inhibitory) neurons, this promoter may be a safe choice to drive ATP1A3 expression in clinically relevant neurons of the AHC mice.
  • Ribosome Binding Site
  • In certain embodiments, the nucleic acid regulatory sequence comprises a sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene. For instance, a Kozak sequence is included to maximize the ribosome binding and translation efficiency of the supplied transgenic ATP1A3.
  • In certain embodiments, GCCACC sequence was included. This sequence represents the most common conserved RBS (ribosome binding site). In certain embodiments, aCCGGAgccacc sequence (SEQ ID NO: 7) was included. In one embodiment, the sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene is included at the 3′ end of the promoters immediately in front of the ATG start codon of the subsequence ATP1A3 ORF.
  • 3′ UTR and Poly(A) Signal
  • The non-coding region 3′ to the protein-coding sequence may be retained for its transcriptional termination regulatory sequences, such as those which provide for termination and polyadenlylation. By retaining the 3′-region naturally contiguous to the coding sequence, the transcriptional termination signals may be provided. Where the transcriptional termination signals natively associated with the coding sequence are not satisfactorily functional in the expression host cell, then a different 3′ region, functional in the host cell, may be substituted.
  • In certain embodiments, the nucleic acid regulatory sequence comprises a 3′-UTR sequence that contains a polyadenylation sequence. In one embodiment, the 3′-UTR sequence is a rabbit beta globin polyadenylation sequence (rBGpA). For instance, 127-bp rabbit beta globin polyadenylation (rBGpA) sequence is employed as the 3′UTR due to its small size and proven effectiveness.
  • In certain embodiments, the recombinant AAV vector comprises an AAV vector, a PATP1A3 promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a PATP1A3 promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • In one embodiment, the recombinant AAV vector comprises AAV9/PATP1A3-ATP1A3 cDNA-rBGpA.
  • In one embodiment, the recombinant AAV vector comprises SEQ ID NO: 3.
  • In certain embodiments, the recombinant AAV vector comprises an AAV vector, a PhSyn promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a PhSyn promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • In one embodiment, the recombinant AAV vector comprises AAV9/PhSyn-ATP1A3 cDNA-rBGpA.
  • In one embodiment, the recombinant AAV vector comprises SEQ ID NO: 4.
  • In certain embodiments, the recombinant AAV vector comprises an AAV vector, a PCBh promoter, and a ATP1A3 cDNA. In certain embodiments, the recombinant AAV vector comprises an AAV9 vector, a PCBh promoter, a ATP1A3 cDNA, a sequence to regulate ribosome binding and/or translation efficiency, and a 3′-UTR sequence that contains a polyadenylation sequence.
  • In one embodiment, the recombinant AAV vector comprises AAV9/PCBh-ATP1A3 cDNA-rBGpA.
  • In one embodiment, the recombinant AAV vector comprises SEQ ID NO: 5.
  • Another aspect of the invention relates to a method for treating a subject having a neurological disorder associated with mutations in the ATP1A3 gene. The method comprises administering into the central nervous system (CNS) of the subject a recombinant adeno-associated virus (AAV) vector comprising a nucleic acid sequence encoding a ATP1A3 protein, thereby restoring some degree of neurological function in said subject.
  • Mutations in the ATP1A3 gene alone account for nine different neurological disorders. The neurological disorder may be selected from the group consisting of alternating hemiplegia of childhood (AHC); rapid-onset dystonia-parkinsonism (RDP); cerebellar ataxia, areflexia, pes cavus, optic atrophy, and sensorineural hearing loss (CAPOS); early infantile epileptic encephalopathy (EIEE); relapsing encephalopathy with cerebellar ataxia (RECA); dystonia, dysmorphism, encephalopathy, MRI abnormalities, no hemiplegia (D-DEMO); familial hemiplegic migraine (FHM); childhood onset schizophrenia (COS); childhood rapid onset ataxia (CROA); and auditory neuropathy spectrum disorder (ANSD). All patients with loss-of-function ATP1A3 mutations can benefit directly from the ATP1A3-based gene therapy.
  • The method can restore alpha-3-specific NaK-ATPase activity in the central nervous system, thereby restoring some degree of neurological function in symptomatic mammals (animals or humans) with mutations in ATP1A3 gene. The method can restore ATP1A3 function in GABAergic neurons in the CNS, which includes Purkinje neurons of the cerebellum.
  • Another aspect of the invention relates to a method for delivering to the central nervous system of a mammal, a ATP1A3 DNA that is expressed in a cell of the central nervous system. The method comprises administering into the central nervous system of the mammalian a recombinant AAV vector comprising a nucleic acid sequence encoding a ATP1A3 protein, wherein said vector transduces the cell.
  • A “subject,” “individual,” or “patient” is used interchangeably herein, which refers to a vertebrate, e.g., a mammal. Mammals include, but are not limited to, mice, rats, simians, humans, farm animals, sport animals, and pets. In certain embodiments, the subject is human.
  • In certain embodiments, the intended developmental stage (age) of the administration into the subject is neonate, pediatric, or juvenile (pre-adult).
  • All the descriptions and embodiments relating to the recombinant AAV vectors, including various AAV vectors, various ATP1A3-coding nucleic acid sequences, and various types of regulator sequences (including promoters, sequences to regulate ribosome binding, and 3′-UTR sequences), discussed in the above aspect of the invention relating to a composition, are applicable for these aspects of the invention relating to a method of treatment or a method of delivery.
  • This gene therapy/delivery method has been tested in various available AHC mouse models by injecting ATP1A3 cDNA-carrying AAV capsids into CSF (via intrathecal or intracisternal injection), using a similar method used to rescue adult mouse models of Mucopolysaccharidosis and diabetic neuropathy. This approach provides a safer and more imminently testable approach than genomic editing, and a targeted and rapid path to long-term treatment. Because AAV does not integrate into the genome, the methods described herein also mitigate risks associated with unintended off-target effects of genomic editing.
  • Administration/Delivery
  • In these aspects of the invention relating to a method of treatment or a method of delivery, the recombinant AAV vector may be administered to one or more CNS components selected from the group consisting of cerebrospinal fluid (CSF), cisterna magna (cerebellomedullaris cistern), the spinal cord, brainstem (medulla, pons, and midbrain), hippocampus, cerebellum, diencephalon (thalamus, hypothalamus), and telencephalon (corpus striatum, cerebral cortex, or, within the cortex, the occipital, temporal, parietal or frontal lobes).
  • The recombinant AAV vector may be delivered to one or more of these CNS components via intracisternal, intrathecal, or intracerebroventricular injection.
  • One purpose is to maximize distribution of the recombinant AAV vector throughout neurons of the CNS. The AAV vector injection can be optimized for cerebellar (Purkinje neuron) targeting.
  • Sites of AAV injection can include intravenous, intrathecal (IT), intra-cisterna magna (ICM), intraparenchymal, intraventricular, and intracerebroventricular. Intravenous administration of AAV is associated with severe hepatotoxicity risk, immunological response, and is least efficient in CNS penetration due to the blood-brain barrier (Hinderer et al., 2018). Additionally, both intraventricular and intraparenchymal routes require invasive intracranial surgery, and the latter has shown very limited distribution of transduction (Hardcastle et al., 2018).
  • Administration of recombinant AAV vector directly into the cerebrospinal fluid (CSF) can deliver the vector directly to the CNS (bypassing the blood brain barrier), distribute it around and throughout the nervous system, and result in wide pattern of neuronal transduction without transfecting internal organs (Federici et al., 2012), thus bypassing immunological response and eliminating the risk of hepatic cytotoxicity associated with intravenous administration (Hinderer et al., 2018). CSF delivery of AAV9 is proven safe and highly efficient in targeting the CNS in various mammals, such as adult mice and pigs.
  • Additionally, direct injection into CSF requires 100×lower dosage than intravenous delivery (Guo et al., 2016). Since transduction rates are highest near the site of delivery, injection of the recombinant vector at the level of cisterna magna (cerebellomedullary cistern, ICM) below the cerebellum can ensure direct and maximal exposure of the vector to cerebellar, hippocampal, brain stem, and cortical regions that show high levels of ATP1A3 expression and are responsible for locomotor and memory impairments in AHC patients.
  • The intra-cisterna magna (ICM) route of the AAV injection results in superior CNS distribution of AAV vectors compared with other routes of CSF access (Hinderer et al., 2014).
  • Administration of AAV directly into the CSF fluid through an intrathecal (IT) route circulates AAVs throughout the CNS and results in widespread CNS transduction. For example, IT administration of 2×1011 vg of AAV9 in pigs resulted in broad spinal motor neuron and extensive cerebellum transduction, with undetectable levels of peripheral organ transduction (Federici et al., 2011). Intrathecal delivery of contrast media, anesthetics, chemotherapeutic agents and drugs for dystonia, spasticity or pain is routinely performed in human medicine. Alternatively, even less invasive IT injection into lumbar cistern was sufficient to achieve broad and efficient transfection of the spinal cord, pons, cerebellum and other CNS parts in mice (Guo et al., 2016).
  • Intrathecal injection is desirable for AAV delivery because it is:
      • a) less surgically invasive than intracranial delivery method;
      • b) more direct and efficient for cerebellar targeting compared to an intravenous method;
      • c) less likely to cause peripheral organ toxicity than intravenous method (requires lower 100×lower dosage (Guo et al., 2016));
      • d) familiar to medical personnel and widely practiced in human medicine (for other purposes); and
      • e) already tested and shown effective for gene delivery into CNS in mice, pigs, and non-human primates.
  • The injection dosage for the intracerebroventricular, IT, or IV administration into the CSF fluid of a mammal should be an effective dose, or effective multiple doses, of the composition comprising the recombinant AAV vector. A single dose may range from 1×109 vg (vector genomes)/kg dose to 1×1015 vg/kg, for instance, from 0.5×1010 vg dose to 5×1011 vg/kg, from 1×1010 vg/kg to 5×1011 vg/kg, or from 1×1012 vg/kg to 1×1014 vg/kg. For instance, a single dose injected into a mouse brain (via intracisternal or intraventricular injection) may range from 1×109 vg/kg to 1×1012 vg/kg.
  • In experimental mice, the total volume of injected AAV solution is, for example, between 1 to 20 μl. For other mammals, including the human brain, volumes and delivery rates are appropriately scaled. For example, it has been demonstrated that volumes of 150 μl can be safely injected in the primate brain (Janson et al. (2002) Hunt. Gene Ther. 13:1391-1412). Treatment may consist of a single injection per target site, or may be repeated in one or more target sites. Injections can be single or multiple, unilateral or bilateral.
  • In an illustrative embodiment, experimental results have proven that the gene-coding AAV9 vectors are capable of transducing cerebellum and increasing a protein and activity levels. For instance, an endogenous mouse ATP1A3 promoter (PATP1A3) was used in AAV9 vector, and the results show that expression of mCherry protein in the neurons of ventral cerebellum was detected in mice injected with the AAV9 vector with mouse PATP1A3 promoter driving mCherry. The sequence of such recombinant AAV9 is shown in SEQ ID NO: 2.
  • In another illustrative embodiment, experimental results have proven that the gene-coding AAV9 vectors are capable of transducing brainstem and increasing a protein levels. The results show the presence of mCherry protein in mouse brainstem following injection of a recombinant vector comprising AAV9/PATP1A3-mCherry cDNA-rBGp(A): there were visible quantity of mCherry protein in cross-sections of the six brainstems of injected mice. The sequence of such recombinant AAV9 is shown in SEQ ID NO: 2.
  • These results indicate that the endogenous PATP1A3 promoter-containing AAV9 vector can transduce neurons of the CNS in mouse in vivo. The results also indicate that the PATP1A3 promoter supports protein expression in neurons of the brainstem in vivo after 4 weeks. Additionally, the results suggest that the endogenous PATP1A3 promoter-containing AAV9 vector would be able to drive expression (transcription and translation) of the protein code it carries in Purkinje neurons.
  • The presence of mCherry protein in mouse brainstem following the injection of AAV9/PATP1A3-mCherry cDNA-rBGp(A) is significant. This is because for a transgenic protein to be present in detectable amounts in the tissue, a sequential series of processes have to occur:
      • Membrane receptor binding and internalization of the AAV vector from the extracellular space into the intracellular neuronal space.
      • Synthesis of the second (complementary) strand on the viral DNA (the injected vector's DNA is single-stranded).
      • Transcription: recognition and binding of the expression cassette's regulatory elements by the endogenous transcription machinery, RNA Polymerase and cis-acting transcription factors, in order to synthesize messenger RNA (mRNA) from the DNA.
      • Translation: recognition and binding of the regulatory elements in the mRNA by the endogenous translation machinery (ribosome and associated proteins) to synthesize and fold protein based on the mRNA.
        The successful completion of each of these processes is critical; if any of these processes is not working, there is no protein expression at the end. Each of these processes requires separate elements to initiate, support, and terminate each process. Therefore, the whole design process is very unpredictable, because there are lots of elements for errors and lots of steps where the process can fail.
  • Successful design of a DNA cassette that supports all these processes and that still fits into a AAV9 capsid is therefore significant, because it needs to include not only the coding sequence for the desired protein, but also has all the necessary regulatory elements to support DNA synthesis (second strand) and transcription (mRNA synthesis) and translation processes (protein synthesis and folding).
  • In this illustrative example, assembled together was an exogenous DNA construct from mouse (PATP1A3 promoter), rabbit (rBGp(A) sequence), jellyfish (mCherry), some generic eukaryotic elements (Kozak sequence), and viral AAV2 DNA (ITR elements). To generate such an exogenous DNA construct that would be recognized by the endogenous cellular machinery in the mouse CNS to drive the various processes to produce new protein in vivo in an amount that, when distributed throughout the expressing neuron, would be visible in a cross-section of the brainstem—this was a very challenging task, particularly in combination with the another factor that all these elements of the DNA construct need to fit within very stringent size constraints of the DNA load that an AAV vector can carry.
  • Therefore, this is a surprising success, even if some DNA elements may have been shown to work elsewhere in other constructs. This is true even though AAV9 serotype is known to have affinity for transfecting neurons (as compared to other AAV types), because the cellular specificity of AAV9 transduction and protein expression is very influenced by the promoter it carries in its DNA (Watakabe et al., 2015). In fact, the specific AAV9 assembly of the parts is new and unique and there is no predictability that all the individual parts would work together (be sufficient to do their job without interfering with the others'). It is the combination of specific capsid serotype and the promoter together that results in the transduction efficiency and cellular tropism (specificity) of transgenic construct expression. The risk factor increases when the DNA construct contains untested components (in this case, the endogenous mouse PATP1A3 promoter).
  • EXAMPLES
  • The following examples are given as particular embodiments of the invention and to demonstrate the practice and advantages thereof. It is to be understood that the examples are given by way of illustration and are not intended to limit the specification or the claims that follow in any manner.
  • Exemplary rAAV9 Sequences:
  • An exemplary mouse promoter PATP1A3 sequence
    (SEQ ID NO: 1)
    acatactgcaagatggtggcactctggggccctgcattactgcaattcactgggcctttcctcccaccctgtatctaccccactcccagaa
    ggaggcagattccagggtgcctcaccctcaaagcctcggtccctaagatacctccctatattgagggggggtctctgagtccccaccctgg
    ggatgtccgggatcaccccccccccccgcactgtgctcagcttctcagtggccgccactttgcagaaacaaggttggagcggtgagggggg
    gaagggggagtacagctgcagtactgggggccgggccgcaagctgtccgtctgctcagtactgctcctgattggccggagccgcctcccc
    ccgcgggcgcgggcatatgaggaggcggaggccccggccgccgcAgcctctgtgcggtgggacccacggaccgacagacgcacgc
    tcccaccgcggcgcgggcgctgcagaggcccccagcccgagcccgcgcctgagcccatcctgcggccaccgctcatcagtctgaaccc
    gctcttcccgcgg
    An exemplary AVV9/PATP1A3-mCherry cDNA-rBGpA sequence (SEQ ID NO: 2).
    ITR2-Patpla3-mCherry-rBGp(A)-ITR2
    GCCACC = Kozak sequence
    Cctgcaggcagctgcgcgctcgctcgctcactgaggccgcccgggcaaagcccgggcgtcgggcgacctttggtcgcccggcctcagt
    gagcgagcgagcgcgcagagagggagtggccaactccatcactaggggttcct
    gcggccgcaCGCGT
    acatactgcaagatggtggcactctggggccctgcattactgcaattcactgggcctttcctcccaccctgtatctaccccactcccagaa
    ggaggcagattccagggtgcctcaccctcaaagcctcggtccctaagatacctccctatattgagggggggtctctgagtccccaccctgg
    ggatgtccgggatcaccccccccccccgcactgtgctcagcttctcagtggccgccactttgcagaaacaaggttggagcggtgagggggg
    gaagggggagtacagctgcagtactgggggccgggccgcaagctgtccgtctgctcagtactgctcctgattggccggagccgcctcccc
    ccgcgggcgcgggcatatgaggaggcggaggccccggccgccgcAgcctctgtgcggtgggacccacggaccgacagacgcacgc
    tcccaccgcggcgcgggcgctgcagaggcccccagcccgagcccgcgcctgagcccatcctgcggccaccgctcatcagtctgaacg
    ccgctcttcccgcgg
    ACCGGTGCCACC atggtgagcaagggcgaggaggataacatggccatcatcaaggagttcatgcgcttcaaggtgcacat
    ggagggctccgtgaacggccacgagttcgagatcgagggcgagggcgagggccgcccctacgagggcacccagaccgccaagct
    gaaggtgaccaagggtggccccctgcccttcgcctgggacatcctgtcccctcagttcatgtacggctccaaggcctacgtgaagcacc
    ccgccgacatccccgactacttgaagctgtccttccccgagggcttcaagtgggagcgcgtgatgaacttcgaggacggcggcgtggt
    gaccgtgacccaggactcctccctgcaggacggcgagttcatctacaaggtgaagctgcgcggcaccaacttcccctccgacggccc
    cgtaatgcagaagaagaccatgggctgggaggcctcctccgagcggatgtaccccgaggacggcgccctgaagggcgagatcaag
    cagaggctgaagctgaaggacggcggccactacgacgctgaggtcaagaccacctacaaggccaagaagcccgtgcagctgccc
    ggcgcctacaacgtcaacatcaagttggacatcacctcccacaacgaggactacaccatcgtggaacagtacgaacgcgccgaggg
    ccgccactccaccggcggcatggacgagctgtacaagtaaGTCGACCCGGGCGGCCTCGAGGACGGGGTG
    AACTACGCCTGAGGATCCGATCTTTTTCCCTCTGCCAAAAATTATGGGGACATCATG
    AAGCCCCTTGAGCATCTGACTTCTGGCTAATAAAGGAAATTTATTTTCATTGCAATA
    GTGTGTTGGAATTTTTTGTGTCTCTCACTCGTccggacacgtgcggaccgagcggccgc
    aggaacccctagtgatggagttggccactccctctctgcgcgctcgctcgctcactgaggccgggcgaccaaaggtcgcccgacgcccg
    ggctttgcccgggcggcctcagtgagcgagcgagcgcgcagctgcctgcagg
    An exemplary PATP1A3-ATP1A3 cDNA-rBGpA sequence (SEQ ID NO: 3).
    >Mmu-407+143 Atp1a3 Promoter = 550 bp total
    >mATP1A3 cDNA in capital letters = 3042 bp
    >rBG poly(A) signal = 131 bp
    acatactgcaagatggtggcactctggggccctgcattactgcaattcactgggcctttc
    ctcccaccctgtatctaccccactcccagaaggaggcagattccagggtgcctcaccctc
    aaagcctcggtccctaagatacctccctatattgagggggggtctctgagtccccaccct
    ggggatgtccgggatcaccccccccccccgcactgtgctcagcttctcagtggccgccac
    tttgcagaaacaaggttggagcggtgagggggggaagggggagtacagctgcagtactgg
    gggccgggccgcaagctgtccgtctgctcagtactgctcctgattggccggagccgcctc
    cccccgcgggcgcgggcatatgaggaggcggaggccccggccgccgcAgcctctgtgcgg
    tgggacccacggaccgacagacgcacgctcccaccgcggcgcgggcgctgcagaggcccc
    cagcccgagcccgcgcctgagcccatcctgcggccaccgctcatcagtctgaacgccgct
    cttcccgcgg_RE_gccacc ATGGGGGACAAAAAAGATGACAAGAGCTCGCCCAAGAAGAGCAAGGCCAAAGAGC
    GCCGGGACCTGGATGACCTCAAGAAGGAAGTGGCTATGACAGAGCACAAGATGTCAGTAGAAGAGGTCTGCCGGAAA
    TACAATACTGACTGCGTGCAGGGTCTGACACACAGTAAAGCCCAGGAGATCCTAGCCCGGGATGGGCCTAACGCCCT
    CACACCACCGCCCACCACCCCAGAATGGGTCAAGTTCTGCCGGCAGCTGTTTGGTGGCTTCTCTATCCTGCTGTGGA
    TCGGGGCAATCCTTTGCTTCCTGGCCTATGGCATCCAGGCAGGGACGGAGGATGACCCTTCCGGTGACAATCTGTAC
    CTGGGCATAGTGCTGGCCGCAGTCGTGATCATCACCGGCTGCTTCTCCTACTACCAAGAAGCCAAGAGTTCTAAGAT
    CATGGAGTCCTTCAAGAACATGGTCCCCCAGCAAGCCCTTGTGATCCGGGAAGGTGAAAAGATGCAGGTGAATGCGG
    AGGAGGTGGTGGTCGGGGACCTGGTGGAGATCAAGGGTGGTGACCGGGTGCCAGCTGACCTGCGCATCATCTCGGCC
    CATGGCTGCAAGGTGGACAACTCCTCCCTGACTGGCGAATCTGAGCCTCAGACCCGCTCCCCGGACTGCACACACGA
    CAACCCCCTGGAGACTCGGAACATCACCTTCTTTTCCACCAACTGCGTGGAAGGCACCGCTCGGGGTGTGGTGGTAG
    CCACAGGTGACCGCACCGTCATGGGCCGCATTGCCACCCTGGCCTCGGGCTTGGAGGTGGGCAAGACGCCCATCGCC
    ATTGAGATTGAGCATTTCATCCAGCTCATTACGGGCGTGGCCGTGTTCCTGGGCGTGTCCTTCTTCATCCTCTCTCT
    CATTCTGGGTTACACCTGGCTCGAGGCAGTCATCTTCCTCATCGGCATCATTGTGGCCAATGTCCCAGAGGGGCTGC
    TGGCTACTGTCACGGTGTGTCTGACGCTGACCGCCAAGCGCATGGCTCGCAAGAACTGCCTGGTGAAGAACCTGGAG
    GCGGTGGAGACGCTGGGCTCCACGTCCACCATCTGCTCGGACAAGACCGGCACTCTCACCCAGAACCGCATGACCGT
    CGCCCACATGTGGTTTGACAACCAGATCCACGAGGCAGACACCACAGAGGATCAGTCAGGGACCTCTTTCGACAAGA
    GCTCACACACCTGGGTGGCCCTGTCCCACATCGCCGGGCTCTGCAACCGGGCCGTCTTCAAGGGCGGGCAGGACAAC
    ATCCCAGTACTCAAGAGGGACGTGGCCGGTGATGCCTCCGAGTCTGCCCTGCTTAAGTGCATTGAGCTGTCCTCGGG
    TTCCGTAAAGCTGATGCGAGAACGGAACAAGAAAGTGGCTGAGATTCCGTTCAACTCGACCAACAAATACCAGCTAT
    CCATCCATGAGACTGAGGATCCCAATGACAACCGGTACCTGCTAGTGATGAAGGGCGCCCCCGAACGCATTCTGGAC
    CGCTGTGCCACCATCCTCCTGCAGGGCAAGGAGCAGCCTCTGGATGAGGAGATGAAGGAGGCCTTCCAGAATGCCTA
    CCTGGAGCTTGGTGGCCTGGGCGAGCGTGTGCTGGGTTTCTGCCATTACTACCTGCCTGAGGAACAGTTCCCCAAGG
    GCTTTGCCTTTGACTGTGATGACGTGAACTTCACCACAGACAACCTTTGCTTTGTGGGTCTCATGTCCATGATTGAC
    CCTCCCCGGGCAGCCGTCCCTGACGCTGTGGGCAAATGCCGAAGCGCAGGCATCAAGGTCATCATGGTCACCGGCGA
    TCACCCCATCACTGCCAAGGCCATTGCCAAGGGTGTGGGTATCATCTCTGAGGGTAACGAGACTGTCGAAGACATCG
    CTGCCCGGCTCAACATCCCTGTCAGCCAGGTGAACCCCAGGGATGCCAAAGCCTGTGTGATTCACGGCACCGACCTC
    AAGGACTTCACCTCGGAGCAGATTGACGAGATTCTGCAGAACCACACCGAGATCGTCTTTGCCCGAACCTCCCCTCA
    GCAGAAGCTCATCATCGTGGAGGGCTGTCAGAGACAGGGAGCAATTGTGGCTGTGACTGGCGATGGTGTGAATGACT
    CCCCTGCTCTGAAGAAGGCTGACATCGGGGTGGCCATGGGCATTGCTGGCTCTGATGTCTCTAAGCAGGCTGCCGAC
    ATGATTCTGCTGGATGACAACTTTGCTTCCATTGTCACTGGTGTGGAGGAAGGCCGCCTGATCTTTGACAACCTGAA
    GAAATCCATCGCCTACACTCTGACTAGCAACATCCCTGAGATCACACCCTTCCTGCTCTTCATCATGGCTAACATCC
    CACTGCCTCTTGGCACCATCACCATCCTCTGCATTGACCTGGGTACCGACATGGTCCCTGCAATCTCCCTGGCCTAC
    GAGGCTGCCGAGAGCGACATCATGAAGAGGCAGCCCAGGAACCCACGCACAGACAAACTGGTCAACGAAAGGCTCAT
    CAGCATGGCCTACGGGCAGATTGGGATGATCCAGGCCCTCGGTGGTTTCTTCTCCTACTTTGTCATCCTGGCAGAAA
    ATGGCTTCTTGCCCGGAAACCTGGTGGGCATCCGGCTCAACTGGGATGATCGCACTGTCAATGACCTAGAAGACAGT
    TATGGGCAGCAGTGGACTTATGAGCAGAGGAAGGTGGTAGAGTTCACATGCCACACAGCCTTCTTTGTGAGTATCGT
    GGTGGTCCAGTGGGCTGACCTGATCATCTGCAAGACCAGGAGGAACTCCGTCTTCCAGCAGGGCATGAAGAATAAGA
    TCTTGATCTTCGGCTTGTTTGAGGAGACGGCCCTCGCTGCCTTCCTGTCCTACTGCCCAGGCATGGATGTGGCCCTT
    CGCATGTACCCTCTCAAGCCCAGCTGGTGGTTCTGTGCCTTCCCCTACAGTTTCCTCATCTTCGTCTATGATGAGAT
    TCGCAAACTCATCCTGCGCAGGAACCCCGGGGGTTGGGTGGAGAAAGAGACCTACTATTGA_RE_gatctttttc
    cctctgccaaaaattatggggacatcatgaagccccttgagcatctgacttctggctaataaaggaaatttattttc
    attgcaatagtgtgttggaattttttgtgtctctcactcggaag
    An exemplary PCBh-ATP1A3 cDNA-rBGpA sequence (SEQ ID NO: 4).
    >CBH promoter + Kozak sequence = 811 bp
    >mATP1A3 cDNA in capital letters = 3042 bp
    >rBG poly(A) signal = 131 bp
    cgttacataacttacggtaaatggcccgcctggctgaccgcccaacgacccccgcccattgacgtcaatagtaacgc
    caatagggactttccattgacgtcaatgggtggagtatttacggtaaactgcccacttggcagtacatcaagtgtat
    catatgccaagtacgccccctattgacgtcaatgacggtaaatggcccgcctggcattgtgcccagtacatgacctt
    atgggactttcctacttggcagtacatctacgtattagtcatcgctattaccatggtcgaggtgagccccacgttct
    gcttcactctccccatctcccccccctccccacccccaattttgtatttatttattttttaattattttgtgcagcg
    atgggggcggggggggggggggggcgcgcgccaggcggggggggcggggcgaggggggggcggggcgaggcggag
    aggtgcggcggcagccaatcagagcggcgcgctccgaaagtttccttttatggcgaggcggcggcggcggcggccct
    ataaaaagcgaagcgcgcggggggggagtcgctgcgacgctgccttcgccccgtgccccgctccgccgccgcctc
    gcgccgcccgccccggctctgactgaccgcgttactcccacaggtgagcggggggacggcccttctcctccgggct
    gtaattagctgagcaagaggtaagggtttaagggatggttggttggtggggtattaatgtttaattacctggagcac
    ctgcctgaaatcactttttttcaggttggaccggt_RE_gccaccATGGGGGACAAAAAAGATGACAAGAGCTCG
    CCCAAGAAGAGCAAGGCCAAAGAGCGCCGGGACCTGGATGACCTCAAGAAGGAAGTGGCTATGACAGAGCACAAGAT
    GTCAGTAGAAGAGGTCTGCCGGAAATACAATACTGACTGCGTGCAGGGTCTGACACACAGTAAAGCCCAGGAGATCC
    TAGCCCGGGATGGGCCTAACGCCCTCACACCACCGCCCACCACCCCAGAATGGGTCAAGTTCTGCCGGCAGCTGTTT
    GGTGGCTTCTCTATCCTGCTGTGGATCGGGGCAATCCTTTGCTTCCTGGCCTATGGCATCCAGGCAGGGACGGAGGA
    TGACCCTTCCGGTGACAATCTGTACCTGGGCATAGTGCTGGCCGCAGTCGTGATCATCACCGGCTGCTTCTCCTACT
    ACCAAGAAGCCAAGAGTTCTAAGATCATGGAGTCCTTCAAGAACATGGTCCCCCAGCAAGCCCTTGTGATCCGGGAA
    GGTGAAAAGATGCAGGTGAATGCGGAGGAGGTGGTGGTCGGGGACCTGGTGGAGATCAAGGGTGGTGACCGGGTGCC
    AGCTGACCTGCGCATCATCTCGGCCCATGGCTGCAAGGTGGACAACTCCTCCCTGACTGGCGAATCTGAGCCTCAGA
    CCCGCTCCCCGGACTGCACACACGACAACCCCCTGGAGACTCGGAACATCACCTTCTTTTCCACCAACTGCGTGGAA
    GGCACCGCTCGGGGTGTGGTGGTAGCCACAGGTGACCGCACCGTCATGGGCCGCATTGCCACCCTGGCCTCGGGCTT
    GGAGGTGGGCAAGACGCCCATCGCCATTGAGATTGAGCATTTCATCCAGCTCATTACGGGCGTGGCCGTGTTCCTGG
    GCGTGTCCTTCTTCATCCTCTCTCTCATTCTGGGTTACACCTGGCTCGAGGCAGTCATCTTCCTCATCGGCATCATT
    GTGGCCAATGTCCCAGAGGGGCTGCTGGCTACTGTCACGGTGTGTCTGACGCTGACCGCCAAGCGCATGGCTCGCAA
    GAACTGCCTGGTGAAGAACCTGGAGGCGGTGGAGACGCTGGGCTCCACGTCCACCATCTGCTCGGACAAGACCGGCA
    CTCTCACCCAGAACCGCATGACCGTCGCCCACATGTGGTTTGACAACCAGATCCACGAGGCAGACACCACAGAGGAT
    CAGTCAGGGACCTCTTTCGACAAGAGCTCACACACCTGGGTGGCCCTGTCCCACATCGCCGGGCTCTGCAACCGGGC
    CGTCTTCAAGGGCGGGCAGGACAACATCCCAGTACTCAAGAGGGACGTGGCCGGTGATGCCTCCGAGTCTGCCCTGC
    TTAAGTGCATTGAGCTGTCCTCGGGTTCCGTAAAGCTGATGCGAGAACGGAACAAGAAAGTGGCTGAGATTCCGTTC
    AACTCGACCAACAAATACCAGCTATCCATCCATGAGACTGAGGATCCCAATGACAACCGGTACCTGCTAGTGATGAA
    GGGCGCCCCCGAACGCATTCTGGACCGCTGTGCCACCATCCTCCTGCAGGGCAAGGAGCAGCCTCTGGATGAGGAGA
    TGAAGGAGGCCTTCCAGAATGCCTACCTGGAGCTTGGTGGCCTGGGCGAGCGTGTGCTGGGTTTCTGCCATTACTAC
    CTGCCTGAGGAACAGTTCCCCAAGGGCTTTGCCTTTGACTGTGATGACGTGAACTTCACCACAGACAACCTTTGCTT
    TGTGGGTCTCATGTCCATGATTGACCCTCCCCGGGCAGCCGTCCCTGACGCTGTGGGCAAATGCCGAAGCGCAGGCA
    TCAAGGTCATCATGGTCACCGGCGATCACCCCATCACTGCCAAGGCCATTGCCAAGGGTGTGGGTATCATCTCTGAG
    GGTAACGAGACTGTCGAAGACATCGCTGCCCGGCTCAACATCCCTGTCAGCCAGGTGAACCCCAGGGATGCCAAAGC
    CTGTGTGATTCACGGCACCGACCTCAAGGACTTCACCTCGGAGCAGATTGACGAGATTCTGCAGAACCACACCGAGA
    TCGTCTTTGCCCGAACCTCCCCTCAGCAGAAGCTCATCATCGTGGAGGGCTGTCAGAGACAGGGAGCAATTGTGGCT
    GTGACTGGCGATGGTGTGAATGACTCCCCTGCTCTGAAGAAGGCTGACATCGGGGTGGCCATGGGCATTGCTGGCTC
    TGATGTCTCTAAGCAGGCTGCCGACATGATTCTGCTGGATGACAACTTTGCTTCCATTGTCACTGGTGTGGAGGAAG
    GCCGCCTGATCTTTGACAACCTGAAGAAATCCATCGCCTACACTCTGACTAGCAACATCCCTGAGATCACACCCTTC
    CTGCTCTTCATCATGGCTAACATCCCACTGCCTCTTGGCACCATCACCATCCTCTGCATTGACCTGGGTACCGACAT
    GGTCCCTGCAATCTCCCTGGCCTACGAGGCTGCCGAGAGCGACATCATGAAGAGGCAGCCCAGGAACCCACGCACAG
    ACAAACTGGTCAACGAAAGGCTCATCAGCATGGCCTACGGGCAGATTGGGATGATCCAGGCCCTCGGTGGTTTCTTC
    TCCTACTTTGTCATCCTGGCAGAAAATGGCTTCTTGCCCGGAAACCTGGTGGGCATCCGGCTCAACTGGGATGATCG
    CACTGTCAATGACCTAGAAGACAGTTATGGGCAGCAGTGGACTTATGAGCAGAGGAAGGTGGTAGAGTTCACATGCC
    ACACAGCCTTCTTTGTGAGTATCGTGGTGGTCCAGTGGGCTGACCTGATCATCTGCAAGACCAGGAGGAACTCCGTC
    TTCCAGCAGGGCATGAAGAATAAGATCTTGATCTTCGGCTTGTTTGAGGAGACGGCCCTCGCTGCCTTCCTGTCCTA
    CTGCCCAGGCATGGATGTGGCCCTTCGCATGTACCCTCTCAAGCCCAGCTGGTGGTTCTGTGCCTTCCCCTACAGTT
    TCCTCATCTTCGTCTATGATGAGATTCGCAAACTCATCCTGCGCAGGAACCCCGGGGGTTGGGTGGAGAAAGAGACC
    TACTATTGA_RE_gatctttttccctctgccaaaaattatggggacatcatgaagccccttgagcatctgacttc
    tggctaataaaggaaatttattttcattgcaatagtgtgttggaattttttgtgtctctcactcggaag
    An exemplary PhSyn-ATP1A3 cDNA-rBGpA sequence (SEQ ID NO: 5).
    >hSyn Promoter = 500 bp total (includes Kozak sequence)
    >mATP1A3 cDNA in capital letters = 3042 bp
    >rBG poly(A) signal = 131 bp
    ttaattaaactagacagactgcagagggccctgcgtatgagtgcaagtgggttttaggaccaggatgaggcggggtg
    ggggtgcctacctgacgaccgaccccgacccactggacaagcacccaacccccattccccaaattgcgcatccccta
    tcagagagggggaggggaaacaggatgcggcgaggcgcgtcgcgactgccagcttcagcaccgcggacagtgccttc
    gcccccgcctggcggcgcgcgccaccgccgcctcagcactgaaggcgcgctgacgtcactcgccggtcccccgcaaa
    ctccccttcccggccaccttggtcgcgtccgcgccgccgccggcccagccggaccgcaccacgcgaggcgcgagata
    ggggggcacgggcgcgaccatctgcgctgcggcgccggcgactcagcgctgcctcagtctgcggtgggcagcggagg
    agtcgtgtcgtgcctgagagcgcagtcgagag_RE_gccaccATGGGGGACAAAAAAGATGACAAGAGCTCGCCC
    AAGAAGAGCAAGGCCAAAGAGCGCCGGGACCTGGATGACCTCAAGAAGGAAGTGGCTATGACAGAGCACAAGATGTC
    AGTAGAAGAGGTCTGCCGGAAATACAATACTGACTGCGTGCAGGGTCTGACACACAGTAAAGCCCAGGAGATCCTAG
    CCCGGGATGGGCCTAACGCCCTCACACCACCGCCCACCACCCCAGAATGGGTCAAGTTCTGCCGGCAGCTGTTTGGT
    GGCTTCTCTATCCTGCTGTGGATCGGGGCAATCCTTTGCTTCCTGGCCTATGGCATCCAGGCAGGGACGGAGGATGA
    CCCTTCCGGTGACAATCTGTACCTGGGCATAGTGCTGGCCGCAGTCGTGATCATCACCGGCTGCTTCTCCTACTACC
    AAGAAGCCAAGAGTTCTAAGATCATGGAGTCCTTCAAGAACATGGTCCCCCAGCAAGCCCTTGTGATCCGGGAAGGT
    GAAAAGATGCAGGTGAATGCGGAGGAGGTGGTGGTCGGGGACCTGGTGGAGATCAAGGGTGGTGACCGGGTGCCAGC
    TGACCTGCGCATCATCTCGGCCCATGGCTGCAAGGTGGACAACTCCTCCCTGACTGGCGAATCTGAGCCTCAGACCC
    GCTCCCCGGACTGCACACACGACAACCCCCTGGAGACTCGGAACATCACCTTCTTTTCCACCAACTGCGTGGAAGGC
    ACCGCTCGGGGTGTGGTGGTAGCCACAGGTGACCGCACCGTCATGGGCCGCATTGCCACCCTGGCCTCGGGCTTGGA
    GGTGGGCAAGACGCCCATCGCCATTGAGATTGAGCATTTCATCCAGCTCATTACGGGCGTGGCCGTGTTCCTGGGCG
    TGTCCTTCTTCATCCTCTCTCTCATTCTGGGTTACACCTGGCTCGAGGCAGTCATCTTCCTCATCGGCATCATTGTG
    GCCAATGTCCCAGAGGGGCTGCTGGCTACTGTCACGGTGTGTCTGACGCTGACCGCCAAGCGCATGGCTCGCAAGAA
    CTGCCTGGTGAAGAACCTGGAGGCGGTGGAGACGCTGGGCTCCACGTCCACCATCTGCTCGGACAAGACCGGCACTC
    TCACCCAGAACCGCATGACCGTCGCCCACATGTGGTTTGACAACCAGATCCACGAGGCAGACACCACAGAGGATCAG
    TCAGGGACCTCTTTCGACAAGAGCTCACACACCTGGGTGGCCCTGTCCCACATCGCCGGGCTCTGCAACCGGGCCGT
    CTTCAAGGGCGGGCAGGACAACATCCCAGTACTCAAGAGGGACGTGGCCGGTGATGCCTCCGAGTCTGCCCTGCTTA
    AGTGCATTGAGCTGTCCTCGGGTTCCGTAAAGCTGATGCGAGAACGGAACAAGAAAGTGGCTGAGATTCCGTTCAAC
    TCGACCAACAAATACCAGCTATCCATCCATGAGACTGAGGATCCCAATGACAACCGGTACCTGCTAGTGATGAAGGG
    CGCCCCCGAACGCATTCTGGACCGCTGTGCCACCATCCTCCTGCAGGGCAAGGAGCAGCCTCTGGATGAGGAGATGA
    AGGAGGCCTTCCAGAATGCCTACCTGGAGCTTGGTGGCCTGGGCGAGCGTGTGCTGGGTTTCTGCCATTACTACCTG
    CCTGAGGAACAGTTCCCCAAGGGCTTTGCCTTTGACTGTGATGACGTGAACTTCACCACAGACAACCTTTGCTTTGT
    GGGTCTCATGTCCATGATTGACCCTCCCCGGGCAGCCGTCCCTGACGCTGTGGGCAAATGCCGAAGCGCAGGCATCA
    AGGTCATCATGGTCACCGGCGATCACCCCATCACTGCCAAGGCCATTGCCAAGGGTGTGGGTATCATCTCTGAGGGT
    AACGAGACTGTCGAAGACATCGCTGCCCGGCTCAACATCCCTGTCAGCCAGGTGAACCCCAGGGATGCCAAAGCCTG
    TGTGATTCACGGCACCGACCTCAAGGACTTCACCTCGGAGCAGATTGACGAGATTCTGCAGAACCACACCGAGATCG
    TCTTTGCCCGAACCTCCCCTCAGCAGAAGCTCATCATCGTGGAGGGCTGTCAGAGACAGGGAGCAATTGTGGCTGTG
    ACTGGCGATGGTGTGAATGACTCCCCTGCTCTGAAGAAGGCTGACATCGGGGTGGCCATGGGCATTGCTGGCTCTGA
    TGTCTCTAAGCAGGCTGCCGACATGATTCTGCTGGATGACAACTTTGCTTCCATTGTCACTGGTGTGGAGGAAGGCC
    GCCTGATCTTTGACAACCTGAAGAAATCCATCGCCTACACTCTGACTAGCAACATCCCTGAGATCACACCCTTCCTG
    CTCTTCATCATGGCTAACATCCCACTGCCTCTTGGCACCATCACCATCCTCTGCATTGACCTGGGTACCGACATGGT
    CCCTGCAATCTCCCTGGCCTACGAGGCTGCCGAGAGCGACATCATGAAGAGGCAGCCCAGGAACCCACGCACAGACA
    AACTGGTCAACGAAAGGCTCATCAGCATGGCCTACGGGCAGATTGGGATGATCCAGGCCCTCGGTGGTTTCTTCTCC
    TACTTTGTCATCCTGGCAGAAAATGGCTTCTTGCCCGGAAACCTGGTGGGCATCCGGCTCAACTGGGATGATCGCAC
    TGTCAATGACCTAGAAGACAGTTATGGGCAGCAGTGGACTTATGAGCAGAGGAAGGTGGTAGAGTTCACATGCCACA
    CAGCCTTCTTTGTGAGTATCGTGGTGGTCCAGTGGGCTGACCTGATCATCTGCAAGACCAGGAGGAACTCCGTCTTC
    CAGCAGGGCATGAAGAATAAGATCTTGATCTTCGGCTTGTTTGAGGAGACGGCCCTCGCTGCCTTCCTGTCCTACTG
    CCCAGGCATGGATGTGGCCCTTCGCATGTACCCTCTCAAGCCCAGCTGGTGGTTCTGTGCCTTCCCCTACAGTTTCC
    TCATCTTCGTCTATGATGAGATTCGCAAACTCATCCTGCGCAGGAACCCCGGGGGTTGGGTGGAGAAAGAGACCTAC
    TATTGA_RE_gatctttttccctctgccaaaaattatggggacatcatgaagccccttgagcatctgacttctgg
    ctaataaaggaaatttattttcattgcaatagtgtgttggaattttttgtgtctctcactcggaag
  • In the above three sequences, the promoter sequence is indicated in lower case letters and marked in boldface. This part of the cassette drives transcription of the transgene and defines the cellular specificity of its expression. In the first sequence, mouse promoter Patp1a3 was used, and may be replaced with the corresponding homologous human sequence for human application, the mouse promoter.
  • Ribosome binding site (=upstream regulatory bit) is indicated in lower case letters and marked in red, and is separated from the promoter sequence by “_RE_”.
  • 3′UTR and poly(A) signal (=downstream regulatory bit) is indicated in lower case letters and marked in green, and is separated from the ATP1A3 protein-coding sequence by “_RE_”.
  • The ATP1A3 protein-coding sequence is the same in all three sequences, indicated in capital letters, and is represented by the open reading frame (ORF) of the most predominant transcript of mouse ATP1A3 gene.
  • For human application, this mouse ATP1A3 protein-coding sequence may be replaced with ORF of the corresponding human sequence, which is also the predominant transcript of human ATP1A3 gene. This human ORF sequence is below (SEQ ID NO: 6).
  • >Hsa_ATP1A3_isoform_1_NM_152296.4
    ATGGGGGACAAGAAAGATGACAAGGACTCACCCAAGAAGAACAAGGGCAAGGAGCGCCGGGACCTGGATG
    ACCTCAAGAAGGAGGTGGCTATGACAGAGCACAAGATGTCAGTGGAAGAGGTCTGCCGGAAATACAACAC
    AGACTGTGTGCAGGGTTTGACCCACAGCAAAGCCCAGGAGATCCTGGCCCGGGATGGGCCTAACGCACTC
    ACGCCACCGCCTACCACCCCAGAGTGGGTCAAGTTTTGCCGGCAGCTCTTCGGGGGCTTCTCCATCCTGC
    TGTGGATCGGGGCTATCCTCTGCTTCCTGGCCTACGGTATCCAGGCGGGCACCGAGGACGACCCCTCTGG
    TGACAACCTGTACCTGGGCATCGTGCTGGCGGCCGTGGTGATCATCACTGGCTGCTTCTCCTACTACCAG
    GAGGCCAAGAGCTCCAAGATCATGGAGTCCTTCAAGAACATGGTGCCCCAGCAAGCCCTGGTGATCCGGG
    AAGGTGAGAAGATGCAGGTGAACGCTGAGGAGGTGGTGGTCGGGGACCTGGTGGAGATCAAGGGTGGAGA
    CCGAGTGCCAGCTGACCTGCGGATCATCTCAGCCCACGGCTGCAAGGTGGACAACTCCTCCCTGACTGGC
    GAATCCGAGCCCCAGACTCGCTCTCCCGACTGCACTCACGACAACCCCTTGGAGACTCGGAACATCACCT
    TCTTTTCCACCAACTGTGTGGAAGGCACGGCTCGGGGCGTGGTGGTGGCCACGGGCGACCGCACTGTCAT
    GGGCCGTATCGCCACCCTGGCATCAGGGCTGGAGGTGGGCAAGACGCCCATCGCCATCGAGATTGAGCAC
    TTCATCCAGCTCATCACCGGCGTGGCTGTCTTCCTGGGTGTCTCCTTCTTCATCCTCTCCCTCATTCTCG
    GATACACCTGGCTTGAGGCTGTCATCTTCCTCATCGGCATCATCGTGGCCAATGTCCCAGAGGGTCTGCT
    GGCCACTGTCACTGTGTGTCTGACGCTGACCGCCAAGCGCATGGCCCGGAAGAACTGCCTGGTGAAGAAC
    CTGGAGGCTGTAGAAACCCTGGGCTCCACGTCCACCATCTGCTCAGATAAGACAGGGACCCTCACTCAGA
    ACCGCATGACAGTCGCCCACATGTGGTTTGACAACCAGATCCACGAGGCTGACACCACTGAGGACCAGTC
    AGGGACCTCATTTGACAAGAGTTCGCACACCTGGGTGGCCCTGTCTCACATCGCTGGGCTCTGCAATCGC
    GCTGTCTTCAAGGGTGGTCAGGACAACATCCCTGTGCTCAAGAGGGATGTGGCTGGGGATGCGTCTGAGT
    CTGCCCTGCTCAAGTGCATCGAGCTGTCCTCTGGCTCCGTGAAGCTGATGCGTGAACGCAACAAGAAAGT
    GGCTGAGATTCCCTTCAATTCCACCAACAAATACCAGCTCTCCATCCATGAGACCGAGGACCCCAACGAC
    AACCGATACCTGCTGGTGATGAAGGGTGCCCCCGAGCGCATCCTGGACCGCTGCTCCACCATCCTGCTAC
    AGGGCAAGGAGCAGCCTCTGGACGAGGAAATGAAGGAGGCCTTCCAGAATGCCTACCTTGAGCTCGGTGG
    CCTGGGCGAGCGCGTGCTTGGTTTCTGCCATTATTACCTGCCCGAGGAGCAGTTCCCCAAGGGCTTTGCC
    TTCGACTGTGATGACGTGAACTTCACCACGGACAACCTCTGCTTTGTGGGCCTCATGTCCATGATCGACC
    CACCCCGGGCAGCCGTCCCTGACGCGGTGGGCAAGTGTCGCAGCGCAGGCATCAAGGTCATCATGGTCAC
    CGGCGATCACCCCATCACGGCCAAGGCCATTGCCAAGGGTGTGGGCATCATCTCTGAGGGCAACGAGACT
    GTGGAGGACATCGCCGCCCGGCTCAACATTCCCGTCAGCCAGGTTAACCCCCGGGATGCCAAGGCCTGCG
    TGATCCACGGCACCGACCTCAAGGACTTCACCTCCGAGCAAATCGACGAGATCCTGCAGAATCACACCGA
    GATCGTCTTCGCCCGCACATCCCCCCAGCAGAAGCTCATCATTGTGGAGGGCTGTCAGAGACAGGGTGCA
    ATTGTGGCTGTGACCGGGGATGGTGTGAACGACTCCCCCGCTCTGAAGAAGGCCGACATTGGGGTGGCCA
    TGGGCATCGCTGGCTCTGACGTCTCCAAGCAGGCAGCTGACATGATCCTGCTGGACGACAACTTTGCCTC
    CATCGTCACAGGGGTGGAGGAGGGCCGCCTGATCTTCGACAACCTAAAGAAGTCCATTGCCTACACCCTG
    ACCAGCAATATCCCGGAGATCACGCCCTTCCTGCTGTTCATCATGGCCAACATCCCGCTGCCCCTGGGCA
    CCATCACCATCCTCTGCATCGATCTGGGCACTGACATGGTCCCTGCCATCTCACTGGCGTACGAGGCTGC
    CGAAAGCGACATCATGAAGAGACAGCCCAGGAACCCGCGGACGGACAAATTGGTCAATGAGAGACTCATC
    AGCATGGCCTACGGGCAGATTGGAATGATCCAGGCTCTCGGTGGCTTCTTCTCTTACTTTGTGATCCTGG
    CAGAAAATGGCTTCTTGCCCGGCAACCTGGTGGGCATCCGGCTGAACTGGGATGACCGCACCGTCAATGA
    CCTGGAAGACAGTTACGGGCAGCAGTGGACATACGAGCAGAGGAAGGTGGTGGAGTTCACCTGCCACACG
    GCCTTCTTTGTGAGCATCGTTGTCGTCCAGTGGGCCGATCTGATCATCTGCAAGACCCGGAGGAACTCGG
    TCTTCCAGCAGGGCATGAAGAACAAGATCCTGATCTTCGGGCTGTTTGAGGAGACGGCCCTGGCTGCCTT
    CCTGTCCTACTGCCCCGGCATGGACGTGGCCCTGCGCATGTACCCTCTCAAGCCCAGCTGGTGGTTCTGT
    GCCTTCCCCTACAGTTTCCTCATCTTCGTCTACGACGAAATCCGCAAACTCATCCTGCGCAGGAACCCAG
    GGGGTTGGGTGGAGAAGGAAACCTACTACTGA
  • Example 1. Experiments Using Wild Type Mice/Neuron Culture
  • Several mouse models of AHC are available for gene therapy testing in vivo. Six distinct mouse strains carrying common AHC-associated ATP1A3 alleles have been generated and characterized. These mouse models phenocopy many clinical symptoms of AHC, demonstrating causality and reproducing the disease severity spectrum observed in AHC patients. These strains thus provide multiple, available, and independent experimental substrates to test the effectiveness of proposed gene delivery in vivo.
  • TABLE 1
    Comparison of clinical features in alternating
    hemiplegia of childhood and ATP1A3 mouse models
    Clinical Mouse models with ATP1A3 missense mutations
    disorder Mashlool Myshkin Matoub
    AHC D801N/+ I810N/+ E815K/+ D801Y/+
    Symptoms
    Hemiplegic episodes + + * +
    Dystonia + + * + +
    Abnormal motor + + + + +
    control/ataxia
    Spontaneous seizures + + + +
    SUDEP + + + +
    Learning and memory + + + + +
    impairments
    Impulsivity/Increased + + + +
    activity in open field
    Abnormal eye +
    movements
    +, feature usually occurs;
    −, feature not described
    * Limb clasping, hindlimb splaying and hindlimb dragging
  • D801N/+ mice were generated via CRISPR-based genomic editing. When introduced into mice, the D801N allele (GAC→AAC) also contains the PAM-blocking silent L8202L mutation (CTG→CTA).
  • I810N/+ frozen embryos were provided by the strain creator, Dr. Steven Clapcote.
  • To determine vector feasibility, mouse primary cerebellar (Purkinje cell) culture is transfected with AAV vector carrying 500 pb mouse ATP1A3 promoter and 3 kb cDNA.
  • Controls include un-transfected neuron culture, mock transfected neuron culture, and neuron culture transfected with AAV with ATP1A3 promoter-driven GFP.
  • Levels of ATP1A3 expression are quantified in each using RT-PCR and Western of lysates.
  • Example 1A. Determining Whether Delivery of Wild Type ATP1A3 Gene Via CSF is Sufficient to Rescue Mouse ATP1A3 KO Model of AHC
  • Inject available KO AHC mice models (DeAndrade, or Ikeda a3+/AE2-6 mice, or Lingrel a3+/KOI4 mice) with ATP1A3 carrying AAV vector. Neonates, 2 week old, and 2 month old mice are tested.
      • Negative control comprises injection of AHC mice with empty AAV vector.
      • Positive control comprises injection of wild type mice at same age, with same vector.
      • Test to measure rescue (Rely on specific known expertise in characterizing the mouse)
      • Follow-up tests: Harvest brains, check ATP1a3 protein and mRNA levels using immunohistochemistry and/or antisense oligo in target brain areas: cerebellum, DCN, brain stem, PV+ interneurons, hippocampus, basal ganglia, thalamus, corpus callosum, and motor cortex.
    Example 1B. Determining Whether Delivery of Wild Type ATP1A3 Gene to CSF is Sufficient to Rescue Mouse ATP1A3 Missense Model of AHC
  • Inject available missense AHC mouse models (D801N, D801Y, I810N and E815K) at site determined in (1) above with ATP1A3 carrying AAV vector. Neonates, 2 week old, and 2 month old mice are tested. The test for rescue is as described for KO mice above.
  • Example 2
  • This example compares specificity and strength of ATP1A3 transgene expression driven by three different AAV9 vectors in mouse brains (AAV vector constructs differ only in their promoter configuration, all the rest of the coding and regulatory DNA sequences are identical among them).
  • Vector biodistribution and expression specificity was determined by double labeling with antibodies against mouse ATP1A3 protein with specific neuronal markers such as Calbindin for Purkinje neurons and GAD67 for GABA-ergic neurons.
  • The first steps consisted of determining the best antibodies and staining conditions to detect ATP1A3 in mouse brain tissues, and in neuronal subtypes: Purkinje cells in the cerebellum and GABAergic neurons in cortex, hippocampus and basal ganglia.
  • 1. Experimental Model
  • Wild type mouse brain embedded in paraffin, sliced at the level of dorsal hippocampus and in the cerebellum.
  • Mice injected with mCherry and ATP1A3 expressing AAV viruses, as shown in Table 1, below.
  • TABLE 1
    Animal Date
    # Sex DOB Treatment Group Injection
    1-1 F Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 26, 2018
    1-2 F Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 26, 2018
    1-3 LL Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 27, 2018
    1-4 F Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 27, 2018
    1-5 M Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 29, 2018
    1-6 M Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 29, 2018
    1-7 M Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 30, 2018
    1-8 M Oct. 30, 2018 AAV9_PATP1A3::mCherry Nov. 30, 2018
    2-1 F Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 26, 2018
    2-2 F Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 26, 2018
    2-3 F Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 27, 2018
    2-4 F Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 27, 2018
    2-5 M Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 29, 2018
    2-6 M Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 29, 2018
    2-7 M Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 30, 2018
    2-8 M Oct. 30, 2018 AAV9_PATP1A3::ATP1A3 Nov. 30, 2018
    3-1 F Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 26, 2018
    3-2 F Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 26, 2018
    3-3 F Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 27, 2018
    3-4 F Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 27, 2018
    3-5 M Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 29, 2018
    3-6 M Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 29, 2018
    3-7 M Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 30, 2018
    3-8 M Oct. 30, 2018 AAV9_PSyn::ATP1A3 Nov. 30, 2018
    4-1 F Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 26, 2018
    4-2 F Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 26, 2018
    4-3 F Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 27, 2018
    4-4 F Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 27, 2018
    4-5 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 29, 2018
    4-6 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 29, 2018
    4-7 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 30, 2018
    4-8 M Oct. 30, 2018 AAV9_PCBh::ATP1A3 Nov. 30, 2018
    1. 8 brains (4m/4f) injected with AAV9_PATP1A3::mCherry
    2. 8 brains (4m/4f) injected with AAV9_PATP1A3::ATP1A3
    3. 8 brains (4m/4f) injected with AAV9_Psynapsin::ATP1A3
    4. 8 brains (4m/4f) injected with AAV9_PCBh::ATP1A3
  • 2. Experimental Procedure and Treatment Schedule
  • Brains received by Novaxia were pre-sliced to isolate the cerebellum, which was dehydrated and included in paraffin blocks. 4 μm slices were obtained from each cerebellum and put on glass slides.
  • Fluofarma performed labelings, starting with multiple test conditions of multiple antibodies:
  • Three ATP1A3 antibodies were tested, all from Rabbits:
      • Sigma/Atlas antibody reference #HPA056446 at 1:1000
      • EMD Millipore antibody reference #06-172-1 at 1:250
      • ProteinTech antibody reference #10868-1-AP at 1:100
  • One Calbindin antibody was tested (from chicken):
      • Synaptic Systems reference #214006 at 1:200
  • Three GAD67 antibodies were tested:
      • Novusbio #NBP1-02161 at 1:1000 and 1:300
      • Synaptic Systems #198 006 at 1:200
      • Millipore #MAB5406 at 1:500
  • One mCherry antibody was tested (from rat):
      • Molecular Probes #M11217 at 1:200
  • ATP1A3 antibodies were tested on hippocampal and cerebellum slices. The calbindin antibody was tested on cerebellum slices and the GAD67 antibody was tested on hippocampal slices.
  • For each antibody, standard immunofluorescence protocols were performed using a Ventana XT automated platform, with two standard antigen retrieval procedures, at pH 6 or pH 8 to determine the best antigen retrieval condition.
  • For ATP1A3, a secondary anti-rabbit Alexa 488 antibody was used for detection while for Calbindin and GAD67 and anti-chicken Alexa 594 antibody was used (Invitrogen, 1:400 dilution).
  • For mCherry detection, as mCherry is a red fluorescent protein, an Alexa 594 anti-rat antibody was chosen for mCherry (1:400 dilution).
  • Additional slides were labeled without the presence of primary antibody to control for potential background/aspecific stainings.
  • For labeling mouse cerebella which were injected with AAV viruses, antigen retrieval was performed at pH8 for all antibodies, and slides were labeled with the anti-mCherry antibody (Molecular Probes #M11217) at 1:200+anti-Calbindin antibody (Synaptic Systems #214006) at 1:200, and anti-ATP1A3 antibody (EMD Millipore antibody #06-172-1) at 1:250+anti-Calbindin antibody (Synaptic Systems #214006) at 1:200.
  • Secondary antibodies were Goat anti-rat Alexa 594 (Invitrogen #A11007)+Goat anti-Chicken Alexa 488 (Invitrogen #A11039) for mCherry/Calbindin and Goat anti-rabbit Alexa 488 (Invitrogen #A11008)+Goat anti-Chicken Alexa 594 (Invitrogen #A11042) for ATP1A3/Calbindin, all at 1:400 dilution.
  • One slice per animal was stained.
  • 3. Results for Staining Development
  • Staining involved the following categories of antibodies: ATP1A3 antibodies, Calbindin antibodies, GAD67 antibodies, and mCherry antibodies.
  • ATP1A3 Antibody: Millipore #06-172-1
  • FIG. 1 depicts ATP1A3 (Millipore #06-172-1) staining in a mouse cerebellum. This ATP1A3 elicitates a good specific signal compared to the signal obtained in the absence of primary antibody, at both antigen retrieval pH. However, a higher signal was obtained with antigen retrieval at pH8. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 2 depicts ATP1A3 (Millipore #06-172-1) staining in mouse cerebellum. The zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition, showing the widespread labeling and higher labeling fluorescence in what appears to be Purkinje cells.
  • FIG. 3 depicts ATP1A3 (Millipore #06-172-1) staining in mouse brain at dorsal hippocampus level. This ATP1A3 antibody elicitates a good specific signal compared to the signal obtained in the absence of primary antibody, at both antigen retrieval pH. However, a higher signal was obtained with antigen retrieval at pH8. All images were acquired with the same microscope settings (exposure time, image dynamic range).
  • FIG. 4 depicts ATP1A3 (Millipore #06-172-1) staining in mouse brain at dorsal hippocampus level. The zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition, showing the widespread labeling. Some cells appear to have higher perinuclear signals in the parietal cortex and in the hippocampus in CA1, CA2-3 and Dentate Gyrus.
  • FIG. 5 shows the ATP1A3 (Millipore #06-172-1) staining in the dorsal hippocampus. The zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition (pH8 antigen retrieval) in the dorsal hippocampus. Arrows show cells with higher labeling.
  • FIG. 6 shows the ATP1A3 (Millipore #06-172-1) staining in the parietal cortex. The zoomed image shows the ATP1A3 signal (green fluorescence channel) in the best condition (pH8 antigen retrieval), in the parietal cortex. Arrows show examples of cells with higher labeling.
  • Calbindin Antibody: Synaptic Systems #214006
  • FIG. 7 depicts Calbindin (Synaptic Systems #214006) staining in mouse cerebellum. This antibody elicited the best specific signal over the negative control in the pH8 antigen retrieval condition.
  • FIG. 8 depicts Calbindin (Synaptic Systems #214006) staining in mouse cerebellum. The zoomed image shows Purkinje cells labeled by the calbindin antibody using the pH8 antigen retrieval condition.
  • GAD67 Antibody: Synaptic Systems #198 006
  • FIG. 9 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level.
  • FIG. 10 depicts GAD67 (Synaptic Systems #198 006) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol. The zoomed image over the dorsal hippocampus (pH8 antigen retrieval protocol) shows the detection of GABAergic neurons with low fluorescence levels.
  • GAD67 Antibody: Millipore #MAB5406
  • FIG. 11 depicts GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level.
  • FIG. 12 depicts GAD67 (Millipore #MAB5406) staining in mouse brain at dorsal hippocampus level, pH8 antigen retrieval protocol. The zoomed image over the dorsal hippocampus (pH8 antigen retrieval protocol) shows good detection of GABAergic neurons in the hippocampus and in the cortex.
  • mCherry Antibody: Molecular Probes #M11217
  • FIG. 13 depicts mCherry staining in mouse cerebellum. Intense stainings are seen below the fourth ventricle and around this region. In the cerebellum, the staining is more diffuse, but specific staining is observed compared to negative brains (infected with ATP1A3 AAV viruses).
  • FIG. 14 depicts the mouse cerebellum anatomy. VERM, indicating the Vermal regions, includes: LING Lingula (I); CENT Central lobule; CUL Culmen; DEC Declive (VI); FOTU Folium-tuber vermis (VII); PYR Pyramus (VIII); UVU Uvula (IX); and NOD Nodulus (X). HEM, indicating the Hemispheric regions, includes: SIM Simple lobule; AN Ansiform lobule; PRM Paramedian lobule; COPY Copula pyramidis; PFL Paraflocculus; and FL Flocculus. CBN, indicating Cerebellar nuclei, includes: FN Fastigial nucleus; IP Interposed nucleus; DN Dentate nucleus; and VeCB Vestibulocerebellar nucleus.
  • FIG. 15 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition. The zoomed images are of the red channel. Intense stainings are seen below the fourth ventricle and around this region. In the cerebellum, the staining is more diffuse with occasionally a more intense cell that could be a Purkinje cell.
  • FIG. 16 depicts mCherry staining in mouse cerebellum with pH8 antigen retrieval condition. The zoomed images are of the red channel at levels of Purkinje cell layers. Arrows show examples of Purkinje cells.
  • 4. Conclusions for Staining Development
  • The Millipore #06-172-1 antibody demonstrated that it was able to detect the target protein, detecting a similar signal as disclosed in the Bottger et al. (2011) publication, in which the specificity of this antibody was shown.
  • Calbindin was well detecting by the tested antibody, giving signals that can be segmented for Purkinje cell detection. The best condition was to use antigen retrieval at pH8.
  • The GAD67 antibody from Synaptic Systems (#198 006) allowed detecting some GABAergic neurons but with low fluorescence levels over background.
  • The GAD67 antibody from Millipore (#MAB5406) allowed detecting GABAergic neurons with good sensitivity and fluorescence levels and can thus be used for further steps of the study plan.
  • The mCherry antibody allowed detecting positive cells only in the mCherry infected group, with strong signals below the fourth ventricle but also labelings in other regions. Co-labeling with the Calbindin antibody will allow determining if specific signals are measured in Purkinje cells. The signals in Purkinje cells are not as strong as below the fourth ventricle.
  • 5. Cerebellum Staining
  • ATP1A3 and Calbindin Staining in Cerebellum
  • Calbindin signals allowed determining the localization of Purkinje neurons. In these, the ATP1A3 signals were peripheral to cells, so once cells were segmented, ATP1A3 was measured in a ring around the calbindin signal.
  • Cell-by-cell data was fed into the proprietary Cytosurfer software to make cytometric analyses. The mean ATP1A3 fluorescence was plotted against cell count (FIG. 17 ) and the cells from the same groups displayed group by group. The intensity histograms were well superposed. A gate was chosen to detect Purkinje cells with higher ATP1A3 intensity (FIG. 18 ) which should appear if ATP13 intensity is increased. FIGS. 19, 20, and 21 show the detected Purkinje neurons and the localization of cells with higher mean intensity. These cells are anatomically regularly spread and do not present a gradient from the cisterna magna. Moreover, in FIG. 21 , most of the cells detected with increased ATP1A3 are false positive due to creases in the cerebellum slice which induces a higher green fluorescence signal.
  • Quantification of ATP1A3 in the different experimental groups shows that the use of ATP1A3 expressing AAV viruses did not increase ATP1A3 mean fluorescence in Purkinje neurons, nor the percentage of cells with increased ATP1A3 expression (FIG. 22 ). Similarly, no differences appeared in other cells that were analyzed (FIG. 23 ).
  • mCherry and Calbindin Staining in Cerebellum
  • mCherry expression was used as a positive control of AAV injection and transgene expression. Cytometric analysis of mCherry intensity in Purkinje neurons showed a similar profile in mCherry infected mice compared to ATP1A3 infected mice (FIG. 24 ). A few cells with higher mCherry fluorescence were detected, but this was artificial as these higher signals appeared where creases in the tissue were present (FIG. 25 ). Quantifications support the absence of differences between treatment groups in Purkinje Neurons (FIG. 26 ) and other cells (FIG. 27 ).
  • 6. Brainstem Staining
  • mCherry Staining in Brainstem
  • Depending on the slices, the brainstem was more or less preserved. mCherry staining was well detected in some brainstems of mCherry-AAV infected brains while no specific signals were present in ATP1A3-AAV infected brains. The pattern of mCherry expression was not widespread but instead focalized in discrete regions, which suggests that the injection site was not truly in the Cisterna Magna but rather in the brainstem itself. Moreover, in some brainstems, no mCherry expression could be detected (FIG. 28 ).
  • ATP1A3 Staining in Brainstem
  • Contrary to mCherry, there were no obvious localization of higher ATP1A3 signals in the brainstem. A few cells showed higher intensity but were spread in the brainstem. The staining intensity in general and staining intensity in highest intensity cells were not different for the different experimental groups and compared to the mCherry-infected group (FIG. 29 ). The interimage variability was higher than the intra image variability and quantifications do not show clear cut differences between ATP1A3 expressing AAV viruses (FIG. 30 ).
  • 7. Data Cerebellum Analysis:
  • Percentage Purkinje neurons with High ATP1A3
    AAV9_ AAV9_ AAV9_ AAV9_
    PAT P1A3:: PAT P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    9.08 0.534 8.91 13.3
    0.957 0.212 0 2.4
    1.19 9.57 10.6 0.742
    0.668 1.39 5.05 1.52
    0.838 3.56 1.77 0.775
    0.175 1.72 0.114 0.644
    2.21 14.4 1.86 0.204
    9.08 1.36 0.629 11.9
  • Mean ATP1A3 fluorescence in Purkinje Neurons
    AAV9_ AAV9_ AAV9_ AAV9_
    PAT P1A3:: PAT P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    99.8 76.2 89.9 98.7
    74.4 73.9 67.8 86.4
    76.7 96 85.4 81.5
    75.9 82 78.3 82.1
    70.3 88 87.1 75.3
    75.2 86.1 71.2 81.5
    85.2 100 84.4 71
    97.9 86.8 72.3 102
  • Mean ATP1A3 fluorescence in other cells
    AAV9_ AAV9_ AAV9_ AAV9_
    PAT P1A3:: PAT P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    80.7 43.7 58.2 71.6
    38.6 38.9 22.5 64.7
    42.9 74.6 47.1 55.9
    40.3 55.2 40.4 56.9
    27.8 67.4 66.8 37.3
    41.1 62.2 26.6 58.6
    66.3 70.2 59.6 26.7
    75.5 67.5 30.6 81.7
  • Percentage Purkinje neurons with High mCherry
    AAV9_PAT ATP1A3
    P1A3:: infected
    mCherry brains
    2.14 0.358
    2.08 5.27
    1.93 2.43
    0.303 3.66
    0 1.44
    1.44 0.149
    1.11
    0.366
  • Mean mCherry fluorescence in Purkinje Neurons
    AAV9_PAT ATP1A3
    P1A3:: infected
    mCherry brains
    16.3 13.5
    13.4 19
    15.1 18.6
    13.8 17.1
    14 14.5
    13.4 12.7
    13
    13.2
  • Mean mCherry fluorescence in other cells
    AAV9_PAT ATP1A3
    P1A3:: infected
    mCherry brains
    127 98.6
    123 132
    93.9 106
    164 118
    113 90.2
    93.3 86.9
    149
    123
  • Brainstem Analysis:
  • Number of mCherry positive cells in brainstem
    ATP1A3 mCherry mCherry
    infected infected infected
    brains brains brains, no Ab
    1 9 0
    1 74 0
    21 26 0
    1 16 0
    6 4 0
    4 29 3
    0 0
    0 0
  • Mean ATP1A3 fluorescence in all cells
    AAV9_PAT AAV9_PAT AAV9_ AAV9
    P1A3:: P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    0.9594657 1.5719883 1.6757175 1.2800999
    1.2781088 1.3089229 1.0601813 1.5957804
    1.3683764 1.9546749 1.2545795 1.3809592
    1.7401803 1.2677235 1.3345006 1.0735391
    0.9455498 1.3787239 1.2546315 1.1570685
    1.7301957 1.3529171 1.3236297 1.4783155
    1.3649146 1.0431079 1.2071395 1.192403
    1.6091142 0.919077 1.1412062 1.3906967
  • Percentage of cells with higher ATP1A3 expression
    AAV9_PAT AAV9_PAT AAV9_ AAV9
    P1A3:: P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    0 0.0141583 0.0179856 22.941409
    13.759039 14.096621 25.746196 0.0182615
    0.0386548 0 21.581866 13.021047
    0.066357 28.1575 13.068149 26.281911
    0.0786164 31.462178 29.967949 25.172961
    0 0 14.827064 0.067128
    0 28.490776 28.233045 21.261966
    0 0 28.181818 15.526353
  • ATP1A3 mean fluorescence in subpopulation with higher ATP1A3
    AAV9_PAT AAV9_PAT AAV9_ AAV9
    P1A3:: P1A3:: Psynapsin:: PCBh::
    mCherry ATP1A3 ATP1A3 ATP1A3
    0 12 12 6.579797
    5.997402 5.9995585 7.1953383 8.75
    5.848485 0 6.5679874 6.0148131
    8.8166668 7.1851386 6.0072859 7.1872419
    10 7.1648367 7.183503 7.1800464
    0 0 6.0020599 6.1937063
    0 8.5209115 7.1846988 6.5676226
    0 0 7.177844 6.0161244

Claims (15)

We claim:
1. A method for treating a subject having alternating hemiplegia of childhood (AHC), the method comprising:
administering into the central nervous system (CNS) of the subject a recombinant adeno-associated virus (AAV) vector composition comprising a nucleic acid sequence encoding a ATP1A3 protein, and a promoter sequence that renders the expression of the ATP1A3 protein-coding nucleic acid sequence specific to the central nervous system; wherein the AAV vector is AAV9, and wherein the promoter is a human neuron-specific promoter with a strong bias towards inhibitory neuron (PhSyn),
thereby restoring some degree of neurological function in said subject.
2. The method of claim 1, wherein the recombinant AAV vector is administered to one or more CNS components selected from the group consisting of cerebrospinal fluid (CSF), cisterna magna (cerebellomedullaris cistern), cerebral ventricles, the spinal cord, brainstem, hippocampus, cerebellum, cerebral cortex, diencephalon, and telencephalon.
3. The method of claim 1, wherein the administration is via intracisternal, intrathecal, or intracerebroventricular injection.
4. The method of claim 1, wherein the subject is human.
5. The method of claim 1, wherein the recombinant AAV vector composition further comprising one or more nucleic acid regulatory sequences, linked directly or indirectly to the ATP1A3 protein-coding nucleic acid sequence.
6. The method of claim 5, wherein the nucleic acid regulatory sequence comprises a sequence to regulate ribosome binding and/or translation efficiency of the ATP1A3 gene.
7. The method of claim 6, wherein the sequence to regulate ribosome binding and/or translation efficiency is a Kozak sequence.
8. The method of claim 5, wherein the nucleic acid regulatory sequence comprises a 3′-UTR sequence that contains a polyadenylation signal sequence.
9. The method of claim 8, wherein the 3′-UTR sequence is a rabbit beta globin polyadenylation signal sequence (rBGpA).
10. The method of claim 5, wherein the recombinant AAV vector comprises AAV9/PhSyn-ATP1A3 cDNA-rBGpA.
11. The method of claim 1, wherein the AAV9 vector has a single-stranded DNA genome.
12. The method of claim 1, wherein the recombinant AAV vector comprises SEQ ID NO: 3.
13. The method of claim 1, wherein the recombinant AAV vector comprises SEQ ID NO: 4.
14. The method of claim 1, wherein the recombinant AAV vector comprises SEQ ID NO: 5.
15. The method of claim 1, wherein the recombinant AAV vector comprises SEQ ID NO: 6.
US18/449,346 2018-04-09 2023-08-14 Aav-mediated delivery of atp1a3 genes to central nervous system Pending US20230381340A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/449,346 US20230381340A1 (en) 2018-04-09 2023-08-14 Aav-mediated delivery of atp1a3 genes to central nervous system

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862654645P 2018-04-09 2018-04-09
US16/379,440 US11738093B2 (en) 2018-04-09 2019-04-09 AAV-mediated delivery of ATP1A3 genes to central nervous system
US18/449,346 US20230381340A1 (en) 2018-04-09 2023-08-14 Aav-mediated delivery of atp1a3 genes to central nervous system

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/379,440 Continuation US11738093B2 (en) 2018-04-09 2019-04-09 AAV-mediated delivery of ATP1A3 genes to central nervous system

Publications (1)

Publication Number Publication Date
US20230381340A1 true US20230381340A1 (en) 2023-11-30

Family

ID=68163293

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/379,440 Active 2040-12-06 US11738093B2 (en) 2018-04-09 2019-04-09 AAV-mediated delivery of ATP1A3 genes to central nervous system
US18/449,346 Pending US20230381340A1 (en) 2018-04-09 2023-08-14 Aav-mediated delivery of atp1a3 genes to central nervous system

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/379,440 Active 2040-12-06 US11738093B2 (en) 2018-04-09 2019-04-09 AAV-mediated delivery of ATP1A3 genes to central nervous system

Country Status (2)

Country Link
US (2) US11738093B2 (en)
WO (1) WO2019199841A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114072514A (en) 2019-05-14 2022-02-18 杜克大学 Compositions and methods for treating ATPase mediated diseases
GB202001930D0 (en) * 2020-02-12 2020-03-25 Univ Sheffield Gene therapy
EP4281034A1 (en) 2021-01-24 2023-11-29 Forrest, Michael, David Inhibitors of atp synthase - cosmetic and therapeutic uses
CN113652427B (en) * 2021-08-20 2023-08-29 深圳市恩辑生物科技有限公司 Mini promoter pATP1B1 and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012115980A1 (en) * 2011-02-22 2012-08-30 California Institute Of Technology Delivery of proteins using adeno-associated virus (aav) vectors
JP6415977B2 (en) * 2011-04-15 2018-10-31 ジェネラックス・コーポレイションGenelux Corporation Cloned strain of attenuated vaccinia virus and method of using the same
KR20220082100A (en) * 2015-05-15 2022-06-16 리젠츠 오브 더 유니버시티 오브 미네소타 Adeno-associated for therapeutic delivery to central nervous system

Also Published As

Publication number Publication date
US11738093B2 (en) 2023-08-29
US20190358346A1 (en) 2019-11-28
WO2019199841A1 (en) 2019-10-17

Similar Documents

Publication Publication Date Title
US20230381340A1 (en) Aav-mediated delivery of atp1a3 genes to central nervous system
US11975043B2 (en) Gene therapy for neurodegenerative disorders
US20210115473A1 (en) METHOD FOR TRANSDUCING Title of Invention CELLS WITH PRIMARY CILIA
JP2020050660A (en) Gene therapy for spinal cord disorders
EP2121914B1 (en) Mitochondrial nucleic acid delivery systems
CN105408352A (en) Effective delivery of large genes by dual aav vectors
KR20200116550A (en) Capsid-free aav vectors, compositions, and methods for vector production and gene delivery
JP7231922B2 (en) AAV-IDUA vectors for the treatment of MPS-I-associated blindness
US20190002916A1 (en) Hybrid dual recombinant aav vector systems for gene therapy
US11680276B2 (en) Compositions and methods for treating retinal disorders
KR20210049131A (en) Gene Sequence of Recombinant Human Type II Mitochondrial Dynein-Like GTPase and Use thereof
CN108103096A (en) A kind of Xian Xing Before-daybreak diseases gene therapy medicament
US20200261600A1 (en) Method for the treatment or prevention of pain or excessive neuronal activity or epilepsy
US20230227515A1 (en) Optimized gene therapy for targeting muscle in muscle diseases
EP4017871A1 (en) Adeno-associated virus vector delivery of alpha-sarcoglycan and the treatment of muscular dystrophy
US20240067989A1 (en) Compositions and Methods for Treating Retinal Disorders
US11357870B2 (en) Codon optimized REP1 genes and uses thereof
US20230149564A1 (en) Aav-naglu vectors for treatment of mucopolysaccharidosis iiib
WO2024011109A2 (en) Compositions and methods for treatment of achromotopsia
WO2024033837A1 (en) Human cone photoreceptor optogenetic constructs

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION