NZ737000B2 - Non-human animals having a disruption in a c9orf72 locus - Google Patents

Non-human animals having a disruption in a c9orf72 locus Download PDF

Info

Publication number
NZ737000B2
NZ737000B2 NZ737000A NZ73700016A NZ737000B2 NZ 737000 B2 NZ737000 B2 NZ 737000B2 NZ 737000 A NZ737000 A NZ 737000A NZ 73700016 A NZ73700016 A NZ 73700016A NZ 737000 B2 NZ737000 B2 NZ 737000B2
Authority
NZ
New Zealand
Prior art keywords
weeks
mice
cell
cells
rodent
Prior art date
Application number
NZ737000A
Other versions
NZ737000A (en
Inventor
Amanda Atanasio
Guochun Gong
Burcin Ikiz
Fralish Michael L Lacroix
Ka Man Venus Lai
David M Valenzuela
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority claimed from PCT/US2016/034304 external-priority patent/WO2016196185A1/en
Publication of NZ737000A publication Critical patent/NZ737000A/en
Publication of NZ737000B2 publication Critical patent/NZ737000B2/en

Links

Classifications

    • 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
    • A01K2267/0318Animal model for neurodegenerative disease, e.g. non- Alzheimer's
    • 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
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • 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
    • A01K2267/0393Animal model comprising a reporter system for screening tests
    • 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
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/02Breeding vertebrates
    • 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
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells

Abstract

non-human animal model for neurodegenerative and/or inflammatory diseases is provided, which nonhuman animal comprises a disruption in a C9ORF72 locus. In particular, non-human animals described herein comprise a deletion of the coding portion of exon 2 through the coding portion of exon 11 of an endogenous C9ORF72 locus. Methods of identifying therapeutic candidates that may be used to prevent, delay or treat one or more neurodegenerative (e.g., amyotrophic lateral sclerosis (ALS, also referred to as Lou Gehrig's disease) and frontotemporal dementia (FTD)), autoimmune and/or inflammatory diseases (e.g., SLE, glomerulonephritis) are also provided endogenous C9ORF72 locus. Methods of identifying therapeutic candidates that may be used to prevent, delay or treat one or more neurodegenerative (e.g., amyotrophic lateral sclerosis (ALS, also referred to as Lou Gehrig's disease) and frontotemporal dementia (FTD)), autoimmune and/or inflammatory diseases (e.g., SLE, glomerulonephritis) are also provided

Description

/034304 NGN—HUMAN ANIMALS HAVlNG A DISRUP'l‘lON IN A C'QQRF72 LOCUS CRGSS REFERENCE TG RELATED Al’PLlCA’l‘lGNS {dill} This application claims the benelit of priority from US. Provisional Application No. 62/l68,l7l, tiled May 29, 2(ll5, US. Provisional Application No. 62/232,658, filed September 25, 2(ll5, and US. Provisional Application No. 62/245,382, tiled r 23, 20l 5, the entire contents of which are incorporated herein by reference.
ORATEGN BY REFERENCE 0F CE- LISTING [$92] The ce listing in an ASCII text file, named as 32698___l{ll521730l___SequenceListing of 56 id), created on May l9, 2616, and submitted to the United States Patent and Trademark Office via EFS—Weh, is incorporated herein by reference, BACKGRQUND {9&3} Neurodegenerative diseases are major butors to disability and disease. ln particular, amyotrophic lateral sclerosis (ALS, also referred to as Lou Gehrig's disease) and frontotemporal dementia (FTD) are rare nervous system disorders characterized by progressive neuronal loss and/or death. Although aging is viewed as the greatest rislr factor for neurodegenerative e, several genetic components have been discovered, For example, mutations in the zinc superoxide dismutase (SOD!) gene have long been associated with ALS. Also, expanded hexanucleotide repeats of GGGGCC within a non—coding region of the C90RF72 gene have been linked to both ALS and ETD, Currently, there is no cure for either disease, yet treatments that help to manage and/or alleviate symptoms do exist. [tltlél] inflammatory diseases e a vast variety of diseases that are often characterized by genetic mutation(s) that result in an impaired or dysfunctional immune system. Although the mechanisms of, for example, toid arthritis, inflammatory bowl disease and glomerulonephritis are not completely understood, several genetic components have been linked to the various signs and symptoms presented by patients.
Such diseases are characterized lay systemic inflammation and display various abnormalities hout the patient hody. As with ALS and Fill, ents for inflammatory diseases aim only to improve symptoms and slow disease progress. [$95] While various laboratory animal models are extensively used in the development of most eutics, few exist that address neurodegenerative and matory diseases in ways that provide for ation of the exact molecular mechanism by which identified genetic components cause disease. Thus, the manner in which genetic mutations cause neurodegerierative and/or inflammatory disease remains largely unlmown. ideal animal models would contain the same genetic components and represent similar characteristics ofhuman disease. Given the genetic differences between species, there is a high unmet need for the development of improved animal models that closely recapitulate human neurodegenerative and/or inflammatory disease. 0f course, such improved animal models provide significant value in the development of ive therapeutic and/or prophylactic agents.
SEWER/EAR“! liltldfi The present invention encompasses the recognition that it is ble to engineer non—human animals to permit ed in viva systems for fying and developing new therapeutics and, in some ments, therapeutic regimens, which can he used for the treatment rodegenerative diseases, ers and conditions in some embodiments, the in viva systems as described herein can he used for identifying and developing new therapeutics for treating inflammatory diseases, disorders, and conditions, in some embodiments, the in viva s as described herein can also he used for identifying and developing new therapeutics for treating autoimmune diseases, ers, and conditions. Further, non~hurnan animals described herein that comprise a disruption in a C90RF172 locus and/or otherwise functionally silen ted C90RF172 locus, such that a C9ORE72 polypeptide is not expressed or produced, are desirahle, for example, for use in identifying and developing therapeutics that target a GGGGCC hexan ucleotide repeat, C90RF72 transcription and tion, and/or increasing or decreasing levels of CQQRFT’Z, which have been associated with disease in humans. in some embodiments, non~human animals as described herein provide improved in viva systems (or models) for neurodegenerative diseases, disorders and conditions (e.g., ALS and/or ETD). in some embodiments, non—human s described herein provide improved in viva systems (or models) for inflammatory disease, disorders, and conditions. 2016/034304 ltltlfl The present invention provides non—human animal models for amyotrophic l sis (ALS), frontotemporal dementia {FTD}, and/or glomerulonephritis. in various embodiments, non—human animal models for ALS and/or F'l‘l) are provided, which are characterized by a disruption (cg, a deletion of an entire coding region) in a C90RF72 locus. in some embodiments, a disruption in a C9ORF72 locus affects one or more neurons of a nonhuman animal comprising said disruption. in some ernbodii'nents, a disruption in a C901RF72 locus affects one or more of the spleen, cervical lymph nodes, bone marrow, kidney and blood ot‘a non~human animal comprising said disruption. {9&3} In some embodiments, a disruption in a C9013F72 locus of a non-hnman animal as bed herein results in one or more of the ing phenotypes: an ALSn lilee phenotype; splenomegaly; denopathy; glomernlonephritis; an infiltration of one or more of macrophages, monocytes and grannlocytes into the spleen, cervical lymph nodes, bone marrow and/or blood; an infiltration of Fit/80+ macrophages in the kidney and/or liver; a depletion of B and/or T cells in the bone marrow; a decrease of lymphocytes in the blood; and an increase in expression of one or more cytokines (e.g., lL—l7, {Eri l), TNF—cc and Ill—l2) in the serum. {@959} In some embodiments, a disruption (cg, a deletion) in a non—human CQORF72 locus results from an insertion of a nucleic acid sequence that, in some certain embodiments; comprises a reporter gene. [illlllll in some ments, a nonnhuman animal is provided comprising in its genome a deletion of the entire coding ce in a C90RF172 locus, i.e., a deletion of a genomic sequence coding for all CQORFH isofornis (he, isoforms Vl V2 and V3). ll in some embodiments, a deletion is of a genomic segment of about 26 hb in a C90RF72 locus of a nonuhuman animal. in some embodiments, a deletion is of a genomic segment encompassing at least exons 2-l l (e.g., exons 2 —ll of Vi), in whole or in part. in some ments, a deletion includes exons l—l l. ln some embodiments, a (39013572 locus having a deletion comprises a reporter gene. in some embodiments, a reporter gene is operably linked to a C9013F72 promoter. in some ments, a C90RF72 promoter is an endogenous promoter, Will; in some embodiments, a (39012572 locus oi‘a man animal described herein lacks the coding region of enon 2 through the coding region of exon ll, and comprises a reporter gene. ln some embodiments, the reporter gene is operably linlted to a C9GRF72 promoter, in some embodiments, the reporter gene is operably linked to the nonucoding region ofexon 2 (i.e., part of the 5’ UTR) oi‘a C9ORF72 gene, thereby placing the reporter gene in an operable linkage to exon l (he, exon la or exon lb) and the upstream regulatory sequences (including the promoter) of a C90RF72 locus of a nonuhuman animal. In specific embodiments, the operable linkage between a reporter gene and the ding portion of exon 2 is achieved by ed deletion of a 2 genomic sequence from the codon ately after the A'l‘G start codon in exon 2 through the coding region of exon ll, and insertion of a reporter coding sequence without an ATG start codon into the site of the C9ORF72 locus immediately alter the ing ATG start codon in exon 2 of the CQORF72 gene. ln some embodiments, expression ofa reporter gene resembles the expression pattern (or profile) of a CQORFZ? locus. {@913} ln some embodiments, a reporter gene is selected from the group consisting of li-galactosidase (ZacZ), luciferase, green tluorescent protein (GFP), enhanced GFP (eGFP), cyan fluorescent protein (CFP), yellow fluorescent protein (YFP), enhanced yellow fluorescent protein (eYFl’), blue fluorescent protein, (EFF), enhanced blue fluorescent n , DsRed, and, MmGFl’o in some n ments, a reporter gene is ZacZ.
Willi-i} In some embodiments, a non~human animal as described herein is homozygous or heterozygous for a deletion of the entire coding sequence in a C90RP’72 locus, [@915] in some embodiments, a nonuhuman animal as described herein develops one or more phenotypes as described ; in some certain, embodiments, phenotypes are detectable after about 8 weeks of age, {little} In some embodiments, a nonuhuman animal as described herein develops one or more symptoms ofALS and/or FTD during pment; in some certain embodiments, after about 36 weeks of age; in some certain embodiments, alter about 40 weeks of age, ln some embodiments, a non-human animal as described, herein develops progressive motor deticits alter about 36 weeks of age. in some embodiments, a non~ human animal as described herein develops lower motor neuron pathology after about 40 weeks ot‘age. in some ments, a nonnhuman animal as described herein develops a decrease in body weight alter about 36 weeks ofage. in some embodiments, a non— human animal as described herein develops mitochondrial ction in motor neurons during development; in some certain embodiments, mitochondrial dysfunction is characterized by a decrease in one or more of mitochondrial ation, basal respiration, maximal respiration, spare atory capacity, ATP production and proton leak; in some certain embodiments, mitochondrial dyslunction is characterized by an increase in the mitochondrial to nuclear DNA ratio as ed to the mitochondrial to nuclear DNA ratio of the motor neurons from a l or reference non—human animal. [lltll'F’l in some ments, a non~human animal as described herein develops one or more symptoms of glomerulonepliritis during development; in some certain embodiments, after about 35 weeks oi‘age, alter about 35—4l weeks of age inclusive or after about 35—60 weeks of age inclusive. in some embodiments, a non-human animal as described herein develops splenomegaly after about 8 weeks of age, In some embodiments, a non—human animal as bed herein develops lymphadenopathy after about 8 weeks of age. in some embodiments, lymphadenopathy is palpable after about lZ—l 8 weeks of age inclusive or after about l 8-60 weeks of age inclusive. in some embodiments, a non—human animal as bed herein is characterized by an ation of one or more ofplasma cells, monocytes, granulocytes and lid/till+ macrophages; in some certain embodiments, infiltration is detectable alter about 8 weeks of age; in some n embodiments, infiltration is able up to 60 weeks of age, ln some embodiments, a non—human animal as described herein develops an infiltration of lid/86+ macrophages in the kidney and/or liver after about 8 weeks of age. {nails} in some embodiments, a non~human animal as described herein develops an increased serum cytokine level of one or more of ill—10, lL—l 2, lli~l7, lli'Nmy, TWP-Of, and MCPd alter about 8 weeks of age. in some embodiments, a non—human animal as described herein develops an increased serum level ol‘anlZ alter about 8 weeks of age that is about 6—fold or more as compared to a reference or control non—human animal. {cars} In some embodiments, a non~human animal as bed herein develops kidney disease characterized by a thickened basement membrane, cast formation (or liyaline cast formation), immune complex deposition, membranoproliferative glornenrlonephritis, interstitial mononuclear inflammation, ulosclerosis, basophilic tubules, or combinations thereof after about 28—35 weeks of age inclusive, after about 3541 weeks ofage inclusive, or after about 35~6tl weeks of age inclusive.) [6929} In some embodiments, a nonuhuman animal as described herein develops an increased myeloid dendritic cell population in one or more of the spleen, lymph nodes, bone marrow, kidney and blood after about 28—35 weelrs of age inclusive. in some embodiments, a myeloid dendritic cell population is characterized as Cii45’*‘coi nylon l l,c+h/,lllCll+, {$621} in some embodiments, a nonnhuman animal as described herein develops an increased serum level of one or more tibodies alter about 8 weeks of age; in some certain embodiments, after about 28—35 weeks of age inclusive. In some embodiments, a non—human animal as described herein develops an sed serum level of one or more autoantibodies between about 8 weeks to about 60 weeks ol‘age inclusive, in some embodiments, one or more autoantibodies are selected from anti—Rheumatoid Factor (anti—RF) dies, anti~dsDNA antibodies, anti-nuclear antibodies (ANA), anti~Smith (anti~Sm) antibodies, antimCardiolipin antibodies, and combinations thereof. [@922] in some embodiments, a nonuhuman animal as described herein develops an increased level of P4/3ll+ hages in one or more of the spleen, lymph nodes, bone marrow, kidney and blood after about 28—35 weeks of age inclusive. ln some embodiments, Fit/80+ macrophages are characterized as CDl lb+F4/’8G+Lyb(}'. [6923} In some embodiments, a nonuhuman animal as described herein develops an increased T cell tion in one or more of the spleen, lymph nodes, bone marrow, kidney and blood after about 28-35 weeks of age inclusive, in some embodiments, T cells are characterized as CD8“:D44: CD8+CD69+, CDSl‘PDll, CDs’l'lCD44: coalescent or GEEK-PD?) In some ments, a non—human animal as described herein develops an increased regulatory T cell population in the spleen and/or lymph nodes alter about 2865 weeks of age inclusive, and wherein the regulatory T cell population is characterized as CD4+lloxP3lI ln some embodiments, a non—human animal as bed herein ps an increased T follicular helper (Till) cells in the spleen, lymph nodes (ego, cervical lymph nodes or "CLN", and mesenteric lymph nodes or "MLN"), and/or blood after about 26 weeks , and wherein the Tilt cell population is characterized as CD4+CXCR5+CD44+lCOS+PD~l +Bcl-6+. {bind} ln some embodiments, a nonrhuman animal as described herein develops an increased plasma cell population in one or more of the spleen, lymph nodes and bone marrow after about $60 weeks ofage inclusive. in some embodiments, a plasma cell population is characterized as D l "CD138+ or amateuralttaaaom‘coissi } In some embodiments, a non~human animal as described herein develops autoimmune lymphoproliterative syndrome (ALPS) during development. In some embodiments, a non-human animal as described herein develops lupus nephritis during development. In some ments, a non—human animal as described herein develops proliferative glomerulonepliropatliy. In some embodiments, a non—human animal as described herein develops one or more phenotypes associated with systemic lupus erythematosus (SLE) during development, In some embodiments, a nonmliuman animal as described herein develops one or more phenotypes or symptoms selected from the group consisting of elevated autoantibody titers and serum nes, lympliadenopathy, splenomegaly and select expansions of myeloid and lymphoid compartments, or a ation f. In some embodiments, one or more phenotypes or symptoms are observed as early as 8 weeks. in some embodiments, one or more phenotypes or symptoms are observed between about l3 weeks to about '24 weeks inclusive. {llllild} In some embodiments, an isolated non—human cell or tissue of a non—human animal as described herein is provided. in some embodiments, an isolated cell or tissue comprises a C9ORF72 locus as described . In some embodiments, a cell is a neuronal cell or a cell from a al lineage. In some embodiments, a cell is from a. id or myeloid lineage. in some embodiments, a cell is selected from a B cell, dendritie tell, macrophage, monocyte, and a T cell. In some embodiments, a tissue is selected from adipose, bladder, brain, breast, bone marrow, eye, heart, intestine, kidney, liver, lung, lymph node, muscle, pancreas, plasma, serum, slrin, spleen, stomach, thymus, testis, ovum, and a combination f. {tillZ'l’} In some embodiments, an immortalized cell line is provided, which is made from an isolated cell of a non—human animal as described herein. {$628} In some embodiments, a nonhuman embryonic stem cell is provided whose genome ses a CQORF72 locus as described herein or a deletion in a CQORF72 locus as described herein. In some ments, a non-human embryonic stem cell is a rodent embryonic stem cell, In some certain embodiments, a rodent embryonic stem cell is a mouse embryonic stem cell and is from a ll? strain, C573L strain, or a mixture f. In some certain embodiments, a rodent embryonic stem cell is a mouse embryonic stem cell and is a mixture of l29 and C5781, strains {$629} In some embodiments, the use of a non—human embryonic stem cell as described herein is provided to make a genetically modified non—human animal. In some certain embodiments, a nonnhuman embryonic stem cell is a mouse embryonic stem cell and is used to make a mouse comprising a (390531772 locus as described herein. in some certain embodiments, a nonhuman embryonic stem cell is a rat embryonic stem cell and is used to make a rat comprising a C9GRF72 locus as described herein. [@938] in some ments, a nonu‘numan embryo is provided comprising, made from, obtained from, or generated from a non~human embryonic stem cell comprising a C9ORF72 locus as described herein, in some certain embodiments, a non—human embryo is a rodent embryo; in some embodiments, a mouse embryo; in some embodiments, a rat . {midi} in some embodiments, the use of a non-human embryo as described herein is provided to make a genetically modified nonnhuman animal. in some certain embodiments, a non—human embryo is a mouse embryo and is used to make a mouse sing a C90RF172 locus as described herein. in some certain embodiments, a non- human embryo is a rat embryo and is used to make a rat comprising a CQORF72 locus as described herein. [6932} In some embodiments, a nucleic acid uct (or targeting construct, or ing vector) as bed herein is provided. {$633} ln some embodiments, a nucleic acid construct as described herein comprises, trom 5' to 3', a non—human targeting arm sing a polynucleotide that is homologous to a 5' portion of a non—human (c.g., a rodent such as a mouse or a rat) 2 locus, a first recombinase recognition site; a first promoter ly linked to a recombinase gene, a second promoter operably linked to a selectable marker, a second recom’binase recognition site, a reporter gene as bed herein, and a non—human targeting arm comprising a polynucleotidc that is gous to a 3' portion ot‘a non—human (cg, a rodent such as a mouse or a rat) (39013572 locus. in some embodiments, the 3' portion of a non—human CQORF72 locus includes a genomic sequence downstream of the stop codon in exon l l of a non—human (cg, a rodent such as a mouse or a rat) 72 gene, in some embodiments, the 5? portion of a C9GRF72 locus includes a genomic sequence upstream of the start codon in exon 2 of a man (eg, rodent such as mouse or rat) (390531772 gene. in many embodiments, first and second recombinase recognition sites are oriented to direct an excision. in many embodiments, a recombinase gene encodes a recombinase that recognizes first and second recombinase recognition sites. in many embodiments, a first promoter drives expression of the recombinase gene in differentiated cells and does not drive expression of the recombinase gene in erentiated cells. in man3/ embodiments, a first promoter is transcriptionally competent and developmentally ted. {$634} in some embodiments, recombinase recognition sites include Earl), loxfil l, iox2272, 2, ioxfio, iox’fl , ZoxMZ, Zox5l7l, FRT, FRTl l, FRT7l , attp, att, FRT, Dre, rox, or a combination thereof. in some embodiments, a recombinase gene is selected from the group consisting of Cre, Flp (cg, Flpe, Flpo), and lire. in some certain embodiments, first and second inase recognition sites are for (cg, onl’) sites, and a recoinbinase gene encodes a Cre r'econibinase. {9365} In some embodiments, a first promoter is selected from the group consisting ofprotamine (Prot; egg Protl or ProtS), Blimpl, Blimpl (l id) tragrnent), Blimpl (2 kb iragment), Qatari, Gata/l, lgl‘Z, thZ, thS, and Pax3. In some certain embodiments, a first promoter is selected from a promoter that s in Table 2. In some certain embodiments, a first promoter is or comprises SEQ ll) N0zfi, SFQ ll) non or SEQ ll) NOz7, [6936} In some embodiments, a selectable marker is selected from group consisting ofneomycin phosphotransferase (neor), hygromycin B phosphotransferase (hygr), puromycinnN—acetyltransferase (purer), blasticitlin S deaminase (hsr‘), xanthine/gnanine ioribosyl transferase (gpt), and Herpes simplex virus thyrnidine kinase (i—lSV~tl<:). {393?} in some embodiments, a second promoter is selected from the group consisting of an UbC er, Uhi promoter, hClVlV promoter, mCMV promoter, CAGGS promoter, lfi‘Fl promoter, pgkl promoter, ctin promoter, and a ROSAZd er. in some certain embodiments, a selectable marker is neoI and a second promoter is Uhi. {$638} in some embodiments, a method ol‘malcing a man animal is provided Whose genome comprises a on of the entire coding sequence in a C90RF72 locus, the method comprising (a) introducing a nucleic acid sequen to into a non—human embryonic stem cell so that the entire coding sequence in a ’Q locus is deleted, which nucleic acid comprises a polynucleotide that is homologous to the C9QRF72 locus; (b) obtaining a genetically modified nonuhuman embryonic stem cell trom {a}; and (c) creating a non—human animal using the genetically modified non~hun1an embryonic stem cell of (b), in some embodiments, a method ofmaking a non—human animal WO 96185 described herein further comprises a step of ng a non—human animal generated in (c) so that a nonuhuman animal homozygous for a deletion is created. {@939} in some embodiments, a nucleic acid sequence is, comprises, or appears in a nucleic acid construct as described , hi some ments, a nucleic acid sequence comprises one or more selection markers. In some embodiments, a nucleic acid sequence comprises one or more pecific recombination sites. In some embodiments, a nucleic acid sequence comprises a recombinase gene and a selection marker flanked by reeombinase recognition sites, which recombinase recognition sites are oriented to direct an excision. in some embodiments, a nucleic acid sequence tinther comprises a reporter gene that is downstream ol’a selection marker. ln some embodiments, a nucleic acid sequence comprises a recombinase gene that is operably linked to a promoter that drives expression of the recombinase gene in ditlerentiated cells and does not drive expression of the recombinase gene in undifferentiated cells, In some embodiments, a nucleic acid sequence comprises a recombinase gene that is ly linked to a er that is transcriptionally ent and developmentally ted. in some embodiments, a nucleic acid sequence comprises a recombinase gene that is ly linked to a promoter that is or comprises SFQ ll) Nflzfi, SEQ ll.) N0:6 or SEQ ll) N027. [@048] in some embodiments, a method for making a nonmhuman animal whose genome comprises a deletion of the entire coding sequence in a C90RF72 locus is provided, the method comprising modifying the genome of a nonrhuinan animal so that it comprises a deletion of the entire coding sequence in a (79031472 locus, thereby making said man animal. filth—ll} In some embodiments, a nonhuman animal is provided which is obtainable by, generated, from, or produced from a method as described herein. [@942] in some embodiments, a northuman animal model ol‘amyotrophic l sclerosis (ALS) or frontotemporal dementia (FTD) is provided, which non—human animal has a genome comprising a deletion of the entire coding sequence in a CQGRF72 locus, Hill-fit In some embodiments, a non~hunian animal model of ophic lateral sclerosis (ALS) or irontotemporal dementia {Fifi} is ed, which is obtained by a deletion of the entire coding sequence in a C9GRF72 locus, wherein the nonhuman animal develops one or more symptoms ofALS and/or FTD during development, l in some embodiments, a non~human animal model of glomerulonephritis is provided, which nonmhuman animal has a genome comprising a deletion of the entire coding sequence in a 72 locus, {$645} in some embodiments, a nonnlmman animal model of glomerulonephritis is provided, which is obtained by a deletion of the entire coding sequence in a CEORF72 locus, wherein the non-human animal develops one or more symptoms of ulonephritis during development.
Hill-filo; in some embodiments, a non~human animal model of lymplioproliferative disease is provided, which nonuhuman animal has a genome comprising a on of the entire coding sequence in a C90RF172 locus. {$647} in some embodiments, a nonnlmman animal model of lymphoproiiferative disease is ed, which is obtained by a deletion of the entire coding sequence in a C90RF172 locus, wherein the nonhuman animal develops one or more symptoms of immune system dysregulation or dysfunction during development. loll-18E in some embodiments, a method for identifying a eutic candidate for the ent of a disease, er or condition in a non~human animal is provided, the method comprising (a) administering a ate agent to a nonhuman animal whose genome comprises a deletion of the entire coding sequence in a C90RF72 locus; (lo) pertorming one or more assays to determine if the candidate agent has an effect on one or more signs, symptoms and/or conditions associated with the disease, disorder or condition; and (c) identifying the ate agent that has an effect on the one or more signs, symptoms and/or ions associated with the disease, disorder or condition as the therapeutic candidate.
Edddgl in some embodiments, a e, disorder or condition in a non—human animal is a neurodegenerative disease, disorder or condition, ln some embodiments, a disease, disorder or condition in a non—human animal is an inflammatory disease, disorder or condition. In some embodiments, a disease, disorder or condition in a non-human animal is an autoimmune disease, disorder or condition. ltitlfitlfi in some embodiments, a disease, disorder or condition in a non—human animal is autoimmune lymphoproliferative me (ALPS; also known as Canale—Smith syndrome). ln some embodiments, AlPS is characterized lay an increased serum level of anltl, anti—Rheumatoid Factor (anti—RE) antibodies, anti—nuclear antibodies (ANA) or combinations thereof. £9951; in some embodiments, a disease, disorder or condition in a man animal is lupus nephritis. in some embodiments, lupus nephritis is characterized by rnesangeal proliferation and/or expansion. in some embodiments, lupus nephritis is characterized by one or more tubular abnormalities. in some ments, one or more tubular abnormalities are selected from dilatation, cast formation, basophilia, and combinations thereof. £99.52; in some embodiments a disease, disorder or condition in a man animal is Systemic Lupus Erythematosus (SLE). in some embodiments, SLE is characterized by one or more of lymphoid liyperblasia, T cell activation, elevated serum antinuclear antibodies (ANA), and systemic inflammation affecting heart, lungs, liver, skin, , nervous system, and kidneys, £9953; to some embodiments, use of a nonuhuman animal as described herein is provided in the manufacture of a medicament for the treatment of a neurodegenerative e, disorder or condition, £9954; in some embodiments, use of a nonnhuman animal as bed herein is provided in the manufacture of a medicament for the treatment of an inflammatory disease, disorder or condition. £9955; in some embodiments, use ot‘a non~human animal as described herein is provided in the manufacture of a medicament for the treatment of an autoimmune disease, disorder or ion. ; in some embodiments, use of a man animal as bed herein is ed in the manufacture of a medicament for the treatment of a lymphoproiiferative disease, disorder or condition. £9957; In some embodiments, use of a non—human animal as described herein is provided in the manufacture of a medicament for the treatment of autoimmune lymphoprolii‘erative syndrome (ALPS). £9958; in some embodiments, use of a non~human animal as described herein is provided in the manufacture of a medicament for the treatment of lupus nephritis. £9959; in some embodiments, a neurodegenerative disease, er or condition is amyotrophic lateral sclerosis (ALS). In some embodiments, a neurodegenerative disease, disorder or condition is frontotemporal dementia (ETD), in some embodiments, an inflammatory e, disorder or condition is glomerulonephritis. in some embodiments, an autoimmune disease, disorder or condition is giomerulonephritis, autoimmune lymphoproliterative syndrome (ALPS), lupus nephritis or systemic lupus erythematosus (SLE). {sees} in some embodiments, an autoimmune disease, disorder or condition as described herein is characterized by a significant increase in serum autoantibody concentration. in some ments, an autoimmune disease, er or condition as described herein is characterized by a significant increase in the serum level of one or more cytolrines (cg, ill/i 0, ill-l2, lL—l7, TNF—u, etc). [illldl] in some embodiments, a l ymphoproliferative disease, disorder or condition as described herein is characterized by a significant increase in one or more immune cells in one or more of the spleen, bone marrow, lymph node(s), kidney or blood, ln some embodiments, a prolii‘erative disease, disorder or condition as described herein is characterized by lation or dysregulation of one or more lymphocytes. {@962} in s embodiments, a deletion of the entire coding sequence in a C90RF72 locus es deletions as described herein. in various embodiments, a non" human CQORF72 locus includes a nonhuman 6.90RF72 locus as described herein. in various embodiments, a non~human CQORFH locus is a murine 72 locus (cg, a mouse or a rat C905f72 locus). [6963} In various embodiments, one or more phenotypes as described herein is or are as compared to a reference or control. in some embodiments, a reference or control includes a non-human animal having a modification as bed herein, a modification that is different than a modification as described herein, or no modification (cg, a Wild type nonuhuman animal}. [llilddl in various embodiments, a man animal described herein is a rodent; in some embodiments, a mouse; in some ments, a rat. [6965} As used in this application, the terms "about" and ximately" are used as equivalents. Any numerals used in this application with or without about/approximately are meant to cover any normal fluctuations appreciated by one of ordinary ski ll in the relevant art, {@966} Other features, objects, and advantages of non-human animals, cells and methods provided herein are apparent in the detailed ption of certain embodiments that s, it should be understood, however, that the detailed description, while indicating certain embodiments, is given by way of illustration only, not limitation.
Various changes and modifications within the scope of the invention will become apparent to those slriiled in the art from the detailed description.
BREEZE DESCRIPTION OF THE DRA‘WING [6967} The Drawing included herein, which is composed of the toliowing Figures, is for illustration purposes only and not for tion. The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawingts) will he provided by the Office upon request and payment of the necessary fee.
{WES} Figure 1A, top box, shows a. schematic illustration, not to scale, of the three reported mouse C90rf72 transcript isoforms (Vi, V2 and V3) and a targeted on gy for disruption of the mouse C9riigf72 locus. A ing vector was ted that includes a mouse gy arm upstream (or "ml-lU", containing a genomic sequence upstream of and inclusive of the start codon in exon 2 of the mouse CQGRF72 gene), a fuel reporter gene (without the ATG start codon), a self—deleting drug ion te (which includes a neomycin resistance gene, and a Cre reconihinase gene linlred to a mouse protaminc l (Prm I) promoter, flanked by loxP sites), and a mouse homology arm downstream (or "infill", containing a genomic sequence 49 hp downstream of the stop codon of exon l l of the mouse (SEQORF:72 gene). Upon homologous recombination, a mouse genomic region of about 26 kl), including the C90rf72 coding sequence for all predicted mouse (390672 isoforrns (ie, the coding ce heginning fiforn the codon immediately after the ATG start codon in exon 2 ofmouse C90rf72, through exons Boll), intervening introns and 49 hp of the E‘UTR in exon l l e C901ff72), was removed; and the incl reporter gene (without the ATG start codon) was inserted immediately alter the remaining, endogenous ATG start codon of mouse Cilaijfiil The resulting modified mouse C90rf72 locus is depicted in Figure lA, hottorn box. Self—deleting technology was employed to remove the neomycin cassette prior to ypic analysis, leaving the lads? er and one loxl’ site under control of the mouse {790:31‘72 promoter. The modified mouse C9orjf72 locus after the neomycin cassette having been deleted is depicted in Figure 1A, bottom box. The nucleotide sequence of the modified (1992772 locus beginning from inserted 1%.? sequence through the 3' loxP site is set forth in SEQ ll) N0: 8; and the nucleotide sequence of the modified C9orf72 locus beginning from exon la through the 3' UTR is set forth in SEQ lD N0: 9.
{F669} Figure iii shows ’l’AQMANCR) expression analysis of C90}:f72 (top; also known as 3110043021RIK) and M08 liinase tor SB (Mobil); bottom) for wild type (wrt hanger/'- (Het) and cacao?" (no) mice» [6979} Figures ZA—ZL show ALS~like phenotypes ed in wild type (n====9) and C9mf72'/" (n=l l) mice, Figure 2A: exemplary percent survival (y—axis) over time (x- axis, weeks); Figure Eli: ary body weight change (y—axis; in grams) over time (x— axis, weeks); Figure 2C: exemplary mean motor impairment score over time (x~axis, ; Figure 21‘}: exemplary mean tremor score over time (x—axis, weeks); Figure 2E: exemplary mean rigidity score over time sl weeks); Figure 2F: exemplary maximum time at rotarod {y—axis, in seconds) overtime (x—axis, weeks); Figure 2G: exemplary open field locomotor behavior, e.g., immobility (left; y~axis, in seconds) and rearing time (right; y—axis, in s) over time (x~axis, weeks); Figure 2H: exemplary catwalk behavior, e,g.; mean stride length (top left, y—axis, eerttimeters [cm]), interlimh nation (top right) presented as percent regularity index (y—axis) over time (x—axis, weeks), and stance phase (bottom center) presented as mean stand (y—axis, in seconds) over time (x—axis; weeks); Figure 2i: ary images of motor neurons from 60 week old wild type (W’l‘, n=5) and C9ory’72"/' mice (11:5), and exemplary motor neuron count (bottom left), mean area (in flmZ, bottom middle, p<tl.00tll) and cell body area (in number of cells, bottom right) for wild type (WT) and C9mgf72"/' mice; l0 motor s were measured for cell body area per slide (three slides per group), swelling indicated hypoxia and cell damage; Figure 2J2 exemplary percent al overtime (top left), body weight change in grams (top right), mean motor impairment score over time (bottom left), mean tremor score over time (bottom middle), and mean rigidity score over time (hottom right) in 32-60 week Ora, wild type (Cgoijf72+/+; who and crown" (n====l7) mice; Figure 2K: exemplary maximum time at d over time (top left), open field locoi'notor behavior, e.g., immobility over time (top middle) and rearing time over time {top right), eatwalh behavior, e,g,, mean stride length over time (bottom left) and interlim’b coordination presented, as percent regularity index over time (bottom middle), and total distance traveled over time (bottom right) in 32—60 week old wild type (C'9orgf72+’/+; n=l4) and 2"; (ri=l7) mice; Figure 2L; exemplary mean motor impairment score over time (top left), mean tremor score over time (top middle); mean rigidity score over time (top right) and grip strength (in grams of force) in wild type (CQOrflffl), heterozygous lZ‘l’l') and homozygous (CQOif72-fl') mice. Statistical significance was determined using Student's unpaired t—test and, one—way analysis of variance (ANGVA) test. {@971} Figures 3A~3AL show immunophenotyping results measured in wild type mice (n= 24) and, mice having a disruption in a C90igf72 locus (n= 3:4). Figure 3A: exemplary images of dissected female wild type (WT) and £790rf72"/' mice showing enlarged cervical lymph nodes (arrows) in C9073f7241' mice, and spleen weights (right, in grams) in female wild type (WT), C90rf7,2+/‘ (HEY), and {7.902772% (KO) mice at 8 (top row) and l8 (hottom row) weeks; Figure SB: exemplary images of dissected male wild ”I" mice showing enlarged cervical lymph nodes (arrows) in type (WT) and ' C9mf72'/" mice, and spleen weights , in grams) in male wild type (WT), (7.902772% (BET), and 72'/" (KG) mice at Elle (top row) and lit (bottom row) weeks; Figure 3C: exemplary images of 37 week female and 56 week male C9olgf‘72'f" mice showing enlarged cervical lymph nodes (arrows); Figure 3E): exemplary images of ted 30— week old female wild type (top row) and (Sitar/W2"; (middle row) mice showing enlarged, cervical lymph nodes s), and body weights (in grams), spleen weights (in grams) and spleen weight normalized to body weight (as % Body weight) (bottom row); exemplary image of ted spleen (bottom led) from wild type (W'l‘) and C90rf72'fl' (~ ll) mice; Figure 3E: exemplary CBC data with differential showing total white blood count and circulating populations of various immune cell types in 34—38 week old male wild type (WT), CmeW’l-‘A (HET), and C90rf724i' (KO) mice (cell type is indicated above each graph); Figure 3F: exemplary images of sectioned spleen and cervical lymph node tissue from wild, type (WT) and, C905f72" mice at 4x power stained, with hematoxylin and eosin; Figure 3G: exemplary images of sectioned al lymph node tissue from C,90ij72"/' mice at 60x power stained with hematoxylin and eosin (blue arrows: cells with plasmacytoid morphology; yellow arrows: phils; green arrows: macrophage—type cells; red arrow: Mott cell); Figure 3B: exemplary percent positive B cells (CDl lh', CDl lc', CD3, 8220+, CDl 9+) irom spleen, cervical lymph nodes, hone marrow and blood in wild type (W'l') and (390672"; (K0) male mice; Figure 31: exemplary plasma cells at various stages expressing specific cell surface antigens isolated from , cervical lymph nodes, hone marrow and blood of male wild type (WT) and C'9orgf72"/‘ (KG) mice at 9~lll weeks (black bars), 18 weeks (light grey bars) and 57~6tl weeks (dark grey bars); Figure 33: exemplary plasma cells at various stages expressing specific tell e ns isolated from spleen, cervical lymph nodes, bone marrow and, blood of female wild type (WT) and C9mj72'fl (KO) mice at 8 weeks (black bars), l8 weeks (light grey bars) and 36—35 weeks (dark grey hars); Figure 3K: exemplary percent positive myeloid cells at various stages expressing specific cell e antigens isolated from spleen, cervical lymph nodes, hone marrow and blood of male wild type (WT) and 72'/' (KG) mice at 9—lG weeks (black bars), l3 weelrs (light grey bars) and 57—60 weeks (dark grey bars); Figure 3L: exemplary t positive myeloid cells at various stages expressing c cell surface antigens isolated from spleen, cervical lymph nodes, bone marrow and, blood of female wild type (WT) and C,90rf72"/' (KO) mice at 8 weeks (black bars), l8 weeks (light grey bars) and 303:5 weeks (dark grey bars); Figure 3M: exemplary percent positive hage (CD452 CD1 lb+, +, Lde’) cells in spleen, cervical lymph nodes, bone marrow, kidney and blood of 30—35 week female wild type (WT) and C9otgf‘72'f" mice; Figure 3N: ary images of ned spleen from wild type (WT) and (790672"; mice at 4x power stained with various markers (indicated beneath each image); Figure 3G: exemplary images of sectioned, cervical lymph node from wild type (WT) and C9orjf72"/' mice at 4X power stained with various markers (indicated beneath each image; arrow: cells intermittently stained with C01 38); Figure 3F: exemplary images of sectioned spleen and cervical lymph node from wild type (WT) and C902f72lfl mice at 4x and 60x power stained with Flt/St}; Figure 3Q: exemplary total cell counts in spleen, cervical lymph nodes, hone marrow and kidney in wild type (WT) and C,9mf72"’l' mice. Cells were counted using a Cellometer Auto 'l‘d Cell Viability Counter PRlGF‘; to FACS analysis. This was done to calculate total number of cells positive for e markers of interest in addition to presenting the data in percent of cells positive for said markers. As these counts were performed alter red blood cell l ysis, they are also representative of a huge immune infiltration (white blood cells) as total cell counts were increased in null mice compared with wild type; Figure 3E: exemplary percent positive and cell counts ofmyeloid dendritic cells (left; out in“, cm tat, uncut) and Ni: cells ; id, corset) in spleen and bone marrow for wild type (WT) and C90rf72"; mice; Figure 3%: exemplary percent positive (top row) and, total cell count (bottom row) for ($345+ cells in various tissues of wild type (WT) and C9otgf‘72'f" mice; Figure 3T: exemplary percent positive (top row) and. total cell count (bottom row) for (238+ T cells in various tissues of wild type (WT) and C90rf72‘fl mice; Figure 3U: exemplary percent positive (top row) and total cell count (bottom row) for CD4+ T cells in various tissues of wild type (WT) and C90}:f72"" mice; Figure 3V: exemplary percent positive (top row) and total cell count (bottom row) for CDti'ltZ‘Délxl+ T cells in various tissues oi‘wild type (WT) and C905f72'/"' mice; Figure 3W: exemplary percent positive (top row) and total cell count (bottom row) for CD4+CD44+ T cells in various tissues of wild type (WT) and C90ry’72'/‘ mice; Figure 3X: exemplary percent positive {top row) and total cell count (bottom row) for posterior” T cells in various s of wild type (WT) and C905???” mice; Figure 3F: ary percent positive (top row) and total cell count (bottom row) for CD4+CD69+ T cells in various s of wild, type (WT) and, £79orf72"/' mice; Figure 32’): exemplary percent positive (top row) and total cell count (bottom row) for C‘Dr'il‘llill+ T cells in various tissues of wild type (WT) and C90ry’72'/" mice; Figure EBA/i: exemplary percent positive (top row) and, total cell count (bottom row) for CD4+PDl l T cells in various tissues of wild type (WT) and C7905f72“/' mice; Figure 3A3: exemplary percent positive (top row) and total cell count (bottom row) for 15r+li‘ox‘l?3+ T cells in various s of wild type (WT) and (Turf/2% mice; Figure EAC: exemplary percent ve (left in each pair) and total cell count (right in each pair) for «customer+ (top left); 62L+ (bottom left), IQT (top right) and Cllzl'l‘Clle 27’+ (bottom right) T cell populations in spleen of wild type (WT) and C90i’f724; mice; Figure EAT}: ary cytohine panel showing expression of various cytokines in l 8 week old male wild type (WT); C9wj72ll‘ (Bet), and C9r)tgf72"’/‘ (KO) mice (cytokines are indicated above each graph); Figure 3AE: exemplary cytolrine panel showing expression of various cytolrines in 8-58 week old male wild type (WT); Glam/72”" (Het), and CQQWZ'L (KO) mice (cytolrines are indicated, above each graph); Figure 3AF: exemplary cytokine panel showing expression of various nes in 8453 week old female wild type (WT); C90rgf72l/i' t), and (390567?" (KO) mice (cytolrines are indicated above each graph); Figure 3AG: exemplary levels ofblood urea nitrogen (yuaxis, mg/dL); globulin (y—axis, gle) and serum immunoglobulin s; lgG, U/rnL (left); y~axis, lglyl, U/mL (right)) in wild type (WT); (390273”) and C9mgf72ll' mice (blood urea nitrogen and globulin measurement is from 45436 week old male mice; serum lgl‘vl and lgG rheumatoid factor measurement is from 8-58 weel: old, male mice; significant increase in lgG and lgl‘vl RF was observed in C9m3f72'f" mice at all time points starting from 8 weeks of age); Figure 3AH: exemplary levels of lgG and lgM Rheumatoid Factor in female (top row) and male (bottom row) wild, type (WT); C905f72l’l' (H et) and C9rirf72‘/' (KG) mice; serum measurement S—ltl week (male: 7 WT; 2016/034304 Het, 9K0; female: 7 WT, 5 Bet, 8 KO), 18 week (male: 9 WT, r3 Het, 13 KO; female: WT, l2 l-let, l5 K0), 304i week (male: 3WT, 4 l-let, 4 KG; female: l0 WT, 9 Het, 9 K0) and 54—65 week (male: 6W1; 9 liet, 5 KO) old mice; Figure 3A1: exemplary levels of circulating lgG and th (in Eng/mill, y-axis) in female {top row) and male (bottom row) wild type (wry, carafe“ (an) and " (ac) mice; Figure 3a.}; ary levels of circulating autoantiliodies (in U/mL, y—axis) in female, 26354 weeks old, (top row) and male, 26n34 weeks old, m row) wild type (WT), Cliorflf’l' (Bet) and C9ffl§f72fl (KO) mice; Figure 3AK: exemplary images of ned kidney (top) and liver (bottom) from wild type (WT) and CQQWZ’A mice at l00x and 20x power, respectively, stained with hematoxylin and eosin (nan) or Emil/80 via liistocliemistry (F4/80 ll-lC); Figure SAL: exemplary images of sectioned kidney tissue from 3—63 week old wild type (WT) and C9mjf72'li' mice d with nematoxylin and eosin (H&E, top panel), lgG (2” panel from top), lglVl (3rd panel iron: top) and Complement C3 {bottom panel). Statistical significance was ined using Students unpaired t—test and one—way analysis of variance (ANQVA) test. [0072} Figure 4 shows exemplary percentage of live neurons to control (y—axis) in cultured wild type neurons treated with various concentrations oftoxins, Top illustrates mental design as described in Example 4. DA (domoic acid; AMPAfkainate receptor agonist), BMAA (fi-Methylarnino~L—alaninc, l00 uM), MKEGl ilpine, l0 gilt/l; NMDA or antagonist), NBQX (2,3"diliydroxyniinnitron7—sulfamoyl— benzoquinoxaline~2,3—diene, 10 ulvl; Alt/ll’A/kainate receptor antagonist); Glutatl’iione {l 0 uM, antioxidant). Statistical significance was determined using Student's impaired t— test and one-way analysis of variance {AN’OVA) a one—way ANOVA test. Cox, PA. et al.,, 2003, Proc, Nat. Acad, Sci. USA, l00(23): 338043383; lit/lurch, SJ. et al.,, 2004, Proc. Nat. Acad. Sci. USA. l0l(33):l2228~l223l; Cox, PA. et al., 2005, Proc. Nat.
Acad. Sci. USA. l02(l4):5074~5078; Erdner, 11L. et al., 2008, Environmental Health, 7(Suppl, 2):SZ. {0073} Figures SA—SD show measurement of ALS~lilte phenotypes in wild type mice stered ip. injections ofEMMA (n=5) or PBS (control, n=5). Figure 5A: top illustrates experimental design and time points for treatment, body weight measurement, and behavioral studies, bottom shows exemplary body weight change (ynaxis, in grams) over time (it—axis, weeks) in control (black) and BMAA—treated mice (grey); Figure SB: exemplary maximum time at rctared (y-axis, seconds) overtime (x axis, weeks) in control (black) and BMAJ-‘tntreated mice (grey); Figure 5C: exemplary open field tor or, e.g., immobility (left; yuaxis, in seconds) and rearing time (right; y— axis; in seconds) overtime s, weeks) in control (black) and BMAA ~treated mice (grey); Figure Si): exemplary catwalk behavior, egt, mean stride length (top left, ynaxis, centimeters Emmi), interlimh coordination (top right) presented as percent regularity index (y—axis) over time (x-axis, weeks), and stance phase (bottom center) presented as mean stand (y—axis, in seconds) over time (x—axis, weeks) in control (black) and BMAAm treated mice (grey). Statistical significance was determined using Student's unpaired tn test and one—way analysis ofvariance (ANGVA) test. {ears} Figures sauna show measurement of ALS-like phenotypes in wild type and 72'fi mice administered in injections ofEMMA (wild type with users: 3 males, 2 females; knockout with BMAA: 3 males, 3 females) or PBS (wild type l: 4 males, l female; knockout control: 5 males, l female). Figure 6A: top illustrates experimental design and time points for treatment, body weight ement, and behavioral studies, bottom left shows exemplary percent survival (y-axis) over time (x— axis, weeks), bottom right shows exemplary body weight change (yuaxis, in grams) over time (x~axis, weeks); Figure tilt: top lefi shows exemplary mean motor ment score over time (x—axis, , top right shows exemplary mean tremor score over time (x— axis, weeks), bottom shows exemplary mean ty score over time (xi—axis, weeks); Figure 6C: exemplary maximum time at rotarod (y~axis, s) over time (x-axis, weeks); Figure 6D: ary open field loeomotor behavior, e,g., immobility (left; y— axis, in seconds) and rearing time (right; y—axis, in seconds) over time (xnaxis, weeks); Figure :6ng exemplary catwalk or, e.g., mean stride length (top left, y-axis, centimeters tomb, interlimh coordination (top right) ted as percent regularity index (ynaxis) overtime (x—axis, weeks), and stance phase m ) presented as mean stand {y~axis, in seconds) over time (x—axis, weeks). tical significance was determined using Student's unpaired t—test and one—way analysis ol‘varianee (ANOVA) test, £9975) Figure 7 shows the in vitro survival (bottom left) and oxidative stress (hottom right) ofwild type and (39047.24— (knockout) motor neurons treated with EMA/3L, Top, schematic illustration of experimental design; Survival (bottom left) is presented as percentage of live neurons to wild type control (y—axis) in wild type (control) and C9rirf72‘/' (knockout) neurons treated with ltlll mlvl BMAA. Oxidative stress at day l (left of bottom right) and day 7 (right of bottom right) is presented as mean optic density ol‘~485/520 nm fluorescence produced by a RGS—sensitive probe? CellRQX Green (Life logies). Statistical cance was determined using Student's unpaired tutest and onenway analysis of variance (AN(EVA) test.) [@976] Figure 8 shows exemplary al (top left)? oxidative stress ( top middle), mitochondrial to nuclear DNA ratio (top right) and various measurements of mitochondrial firnction {bottom left to right) in wild type (WT) and C901f72"; motor neurons. Survival (top left) is presented as numher of cells as percent to C9015 “72%; tive stress at clay l and day 7 is ted as Reactive Oxygen Species (R08; optical density percent to 6902772”+1); Mitochondrial to nuclear DNA ratio is presented as mean mitochondrial copy numher; Mitochondrial respiration is presented, as oxygen consumption rate calculated as percent to C’Qory‘filfl' lSt measurement; Basal respiration, ATP production, Proton leak, Maximal respiration, Spare respiratory capacity and Non— mitochondrial respiration are presented as oxygen consumption rate ated as percent to C'9orjf72+/+ control» [6977} Figures 9Au§C show progressive glornerulonephropathy in C’Qorf‘fi'fi mice.
Figure 9A shows weighted graphs of histopathological scoring that demonstrate that the most significant renal s observed in null mice are associated with rnembranopr‘oliierative glomerulonephritis. Figure 9B shows individual histopathological scores corresponding to weighted graphs ed in Figure 9A.
Briefly, nae stained kidney sections were blindly scored for categories of renal disease associated, with immune ed glomerulonephropathy: menihranoproliferative glomerulonephritis, interstitial mononuclear inflammation} hyaline cast formation, glomerulosclerosis, and hasophilie s. Score of , i=1'ninimal, 2=mild, 3=moderate and 4=severe All null mice displayed minimal to severe rnembranoproliferative glomerulonephritis with occasional evidence of additional disease categories in more severely affected s. Figure 963 shows urine ACR measurements assayed at 14 week (Figure 9C9 top) and 24 week (Figure 9C, bottom) time points from the same cohort of mice indicate onset of albuminuria in C90rjf72'/' mice with age. zygous mice displayed values comparable to WT consistent with the absence of an observed ype. {$678} Figure it) demonstrates that 'l‘ follicular helper (Till) cells {CD4—l-CXCRS-t-CDAlzl-t-ICGSK-l-PDul+Bcl—6k-t) were significantly increased by percent and total cell count in C905f72+ spleen, CLN, MLN and blood, Graphs represent mean i sent. (*9 E (3.05, MP :- tlill, ”*9 E (Milli by unpaired Students t—test )3, 26 week females, n = 5 per genotype. Elevated ’l‘lh cells were also observed in 72"/' BM that did not reach significance, DEFlNlTIGNS [@979] This invention is not limited to ular methods and experimental conditions described herein, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended, to be limiting, since the scope of the present invention is defined by the claims. {thigh} Unless delined otherwise, all terms and phrases used herein include the meanings that the terms and phrases have attained, in the art, unless the ry is clearly indicated or clearly apparent hunt the context in which the term or phrase is used.
Although any methods and materials similar or equivalent to those described herein can he used in the practice or testing of the t invention, particular methods and materials are now described. All ations mentioned, herein are hereby incorporated by nce. {@981} ”Administration "’ includes the administration of a composition to a subject or system (e.g.,, to a cell, organ, tissue, organism, or relevant component or set of components thereof). Those of ry ski ll will appreciate that route of administration may vary depending, for e, on the t or system to which the composition is being administered, the nature of the ition, the purpose of the administration, etc, For example, in certain embodiments, administration to an animal subject (cg, to a human or a rodent) may be ial {including by bronchial instillation), buccal, enteral, interdermal, intra.~aiteria.l, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, al, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by racheal instillation), transden'nal, vaginal and/or vitreal. in some embodiments, administration may involve intermittent dosing. in some embodiments, administration may involve continuous dosing (e.g,, perfusion) for at least a selected period of time, {@332} "Arnelioration ” includes the prevention, reduction or palliation of a state, or improvement of the state of a t. Amelioration includes, but does not require complete recovery or complete prevention ot‘a disease, disorder or condition (ego, radiation inj ury). [0083} ”Approximately”, as applied to one or more values of interest, includes to a value that is similar to a stated reference value. in certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, l9%, l8%, l7%, l6%, l5%, l4%, l3%, l2%, ll%, l0%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, l %, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the content (except where such number would exceed l00% of a possible value). {0034} "Eioiogicolhr active" includes a characteristic of any agent that has ty in a biological system, in vitro or in viva (eg, in an organism). For instance, an agent that, when present in an organism, has a biological eliect within that sm is considered to be biologically . In particular embodiments, where a protein or polypeptide is biologically active, a portion of that protein or polypeptide that shares at least one biological activity of the protein or ptide is lly referred to as a "biologically " portion. [0085} ”Coinpamblc" includes two or more agents, entities, situations, sets of conditions, etc. that may not be identical to one another but that are sufficiently similar to permit comparison there between so that sions may reasonably be drawn based on differences or rities observed. Those of ordinary skill in the art will understand, in context, what degree of identity is required in any given circumstance for two or more such agents, entities, situations, sets of conditions, etc. to be considered comparable. {0036} "Conservative", when describing a conservative amino acid substitution, includes substitution of an amino acid residue by r amino acid e having a side chain R group with similar chemical ties (cg, charge or hydrophobicity). ln general, a conservative amino acid substitution will not substantially change the functional properties ofinterest ot‘a protein, for e, the ability of a receptor to bind to a ligand. Examples of groups of amino acids that have side chains with similar chemical properties include: aliphatic side chains such as glycine, e, valine, leucine, and isoleucine; aliphatic—hydroxyl side chains such as serine and threonine; amide—containing side chains such as asparagine and glutamine; aromatic side chains such as phenylalanine, ne, and tryptophan; basic side chains such as lysine, arginine, and histidine; acidic side chains such as aspartic acid and glutamic acid; and sulfurncontaining side chains such as ne and methionine. Conservative amino acids substitution groups include, for example, /leucine/isoleucine, phenylalanine/tyresine, lysine/arginine, alanine/valine, glutamate/aspartate, and asparagine/glutamine. in some embodiments, a conservative amino acid substitution can be a substitution of any native residue in a protein with alanine, as used in, for example, alanine scanning mutagenesis. be some embodiments, a conservative substitution is made that has a positive value in the PAR/£1250 log~likelihood matrix disclosed in Gonnet, Girl. et at, l992, Science 25614434445. ln some embodiments, a substitution is a moderately vative substitution wherein the substitution has a nonnegative value in the PAM250 log—likelihood matrix. {$687} "Control" includes the derstood meaning of a "corztrol" being a standard against which results are compared. Typically, controls are used to augment integrity in experiments by isolating variables in order to make a conclusion about such variables, in some embodiments, a control is a reaction or assay that is performed simultaneously with a test reaction or assay to provide a comparator. A "corztrol" also includes a "cmztrol animal. " A "control " may have a ation as described herein, a modification that is different as described herein, or no modification (i.e., a wild type animal). In one experiment, a "test” tie, a variable being tested) is applied. ln a second experiment, the "control," the le being tested is not applied. in some embodiments, a control is a historical control (i.e., of a test or assay performed previously, or an amount or result that is previously , in some embodiments, a control is or comprises a printed or otherwise saved record. A l may be a positive control or a ve control. {@338} "Disruption " es the result ofa homologous recombination event with a DNA molecule leg, with an endogenous homologous sequence such as a gene or gene locus). In some embodiments, a disruption may achieve or represent an insertion, deletion, substitution, replacement, rnissense mutation, or a flame—shift of a DNA sequence(s), or any combination thereof. insertions may include the insertion of entire genes or fragments of genes, e.g., exons, which may be of an origin other than the endogenous sequence {e.g., a heterologous sequence). ln some embodiments, a disruption may increase expression and/or activity of a gene or gene t (e.g., of a protein encoded by a gene). in some ments, a disruption may se expression and/or activity ofa gene or gene t. in some embodiments, a disruption may alter sequence of a gene or an encoded gene product (e.g., an encoded protein). In some embodiments, a disruption may truncate or fragment a gene or an encoded gene product (eg, an encoded protein). In some embodiments, a disruption may extend a gene or an encoded gene product. In some such embodiments, a disruption may achieve assembly ofa fusion n. In some embodiments, a disruption may affect level, but not activity, of a gene or gene product. In some embodiments, a disruption may affect activity, but not level, of a gene or gene product. in some embodiments, a tion may have no icant effect on level of a gene or gene product. in some embodiments, a disruption may have no significant effect on activity ofa gene or gene product. In some embodiments, a disruption may have no significant effect on either level or activity of a gene or gene t. [@939] "Determining", "measuring", "evaluating", "assessing", "assaying" and "analyzing” includes any form urement, and include determining ifan t is present or not. These terms include both quantitative and/or qualitative determinations. Assaying may be relative or absolute. "Assayingfor the presence of“ can be determining the amount of something present and/or determining whether or not it is present or absent. {$698} "Endogenous locus" or "erzdogenous gens“ includes a c locus found in a parent or reference organism prior to introduction ofa disruption, deletion, replacement, alteration, or modification as described herein. In some ments, the endogenous locus has a sequence found in nature. in some embodiments, the endogenous locus is a Wild type locus. in some embodiments, the reference sm is a wild type organism. In some embodiments, the reference organism is an engineered organism. In some embodiments, the reference organism is a laboratory—bred organism (Whether Wild type or engineered). [@991] "Endogenous promoter" es a promoter that is naturally associated, e.g, in a wild type organism, with an nous gene. guess} ”Gene" includes a DNA sequence in a. chromosome that codes for a product (e.g., an RNA t and/or a polypeptide product). in some embodiments, a gene includes coding sequence {i.e., sequence that encodes a particular product). in some embodiments, a gene includes non—coding sequence. In some particular embodiments, a gene may include both coding (cg, exonic) and non—coding (e.g., ic) sequence. in some embodiments, a gene may include one or more regulatory sequences (cg, promoters, enhancers, are.) and/or lntron sequences that, for example, may control or impact one or more aspects of gene expression (e.g., cellutypeuspecilic expression, inducible expression, etc). For the purpose of clarity we note that, as used in the present application, the term "gene" generally refers to a portion of a c acid that encodes a polypeptide; the term may optionally encompass tory sequences, as will he clear from context to those of ry ski ll in the art. This definition is not intended to exclude ation of the term "gene" to non—proteinrcoding expression units but rather to clarify that, in most cases, the term as used in this document refers to a polypeptide" coding nucleic acid. ltlll93l "Hererologous " includes an agent or entity from a different source. For example, when used in reference to a polypeptide, gene, or gene t present in a particular cell or organism, the term clarifies that the relevant polypeptide, gene, or gene product: l) was engineered by the hand ofman; '2) was introduced into the cell or organism (or a precursor i) through the hand of man (cg, via genetic engineering); and/or 3) is not naturally produced by or present in the relevant cell or organism (e.g., the relevant cell type or organism type). "iicieroi’ogous" also includes a polypeptide, gene or gene product that is normally present in a particular native cell or organism, but has been modified, for example, by mutation or placement under the control of nonunaturally associated and, in some embodiments, non—endogenous regulatory elements (eg, a promoter).
{Wild} “Hurt cell " includes a cell into which a nucleic acid or protein has been introduced. Persons of skill upon reading this disclosure will tand, that such terms refer not only to the particular subject cell, but also is used to refer to the progeny of such a cell. e certain modifications i'nay occur in succeeding generations due to either mutation or environmental influences, such y may not, in fact, be cal to the parent cell, but are still included within the scope of the phrase "host cell". In some embodiments, a host cell is or comprises a prokaryotic or otic cell. in general, a host cell is any cell that is suitable for receiving and/or producing a heterologous nucleic acid or protein, less of the Kingdom of life to which the cell is designated. ary cells include those of prokaryotes and eukaryotes {single—cell or multiple— cell), bacterial cells (e.g., strains ofEscherichia coli, Bacillus spp., Streptomyces spp., etc), mycohacteria cells, fungal cells, yeast cells {e.g., Saccltaromyces ision, Scltizosoechommyces pombe, Pichiopasz‘oris, Pickle melhanolz‘cu, etc), plant tells, insect cells {e.g., SE9, SF—Zl, baeuloyiiusninfeeted insect cells, lhichcplusia m", etc.), man animal cells, human cells, or cell fusions such as, for example, hybridomas or quadrontas. in some embodiments, the cell is a human, monkey, ape, hamster, rat, or mouse cell. in some embodiments, the cell is eukaryotic and is selected from the following cells: Cl-lO (cg, (ll-K) Kl, DXBd l CEO, Veggie—Cl-lO), COS (e.g., (305—7), retinal cell, Vere, CVl 293 EBNA, MSR 293, MDCK, Bali, , kidney (eg, l-lEK293, nun), Hera, HepGZ, wras, MRC 5, ColoZGS, nu son's, men, tag, BHKZl), at, Daudi, A43l rmal), CV-l, U937, 313, L cell, C127 cell, SPZ/tl, NS~0, MM’l“ , i cell, BRL 3A cell, l-lTlllSll cell, myeloma tell, tumor cell, and a cell line derived from an aforementioned cell. In some embodiments, the cell comprises one or more Viral genes, cg, a retinal cell that expresses a Viral gene leg, a PER.C6® cell). in some embodiments, a host cell is or comprises an isolated cell. in some embodiments, a host cell is part ol‘a tissue. in some embodiments, a host cell is part of an organism. [0095} “Identity”, in tion with a comparison of sequences, includes identity as determined by a number ot‘dit‘l‘erent algorithms known in the art that can be used to measure nucleotide and/or amino acid sequence identity. In some embodiments, identities as described herein are determined using a ClustalW y. l,83 (slow) alignment employing an open gap penalty of l0.0, an extend gap penalty ot‘0.l, and using a Gonnet similarity matrix (M.ACV’ECT()RTM l0.0.2, MacVector inc, 2008). [0096} ve", ase“. "eliminate? or "reduce" includes indicated values that are relative to a baseline measurement, such as a measurement in the same individual (or animal) prior to initiation of a treatment described herein, or a measurement in a control individual (or ) or le control individuals (or animals) in the absence of the treatment described herein. {0097} "Isolated" includes a substance and/or entity that has been (l) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an mental setting), and/or (2) designed, ed, prepared, and/or manufactured by the hand ofman. lsolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 400/ , about 50%, about 60%, about 70%, about 80%, about 90%, about 9l%, about 92%, about 93%, about 94%, about 95%, about 9 %, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated. in some embodiments, isolated agents are about 30%, about 85%, about 90%, about 9l%, about 92%, about 9 %, about 94%, about 95%, about 96%, about 97%, about 98%, about 990/ or more than about 99% pure. in some embodiments, a substan‘e is "pure" ifit is substantially free of other components, in some embodiments, as will be understood by those skilled in the art, a substance may still be considered "isolated" or even "pure", after having been combined with certain other components such as, for example, one or more carriers or excipients (cg, buffer, solvent, water, etc); in such ments, t isolation or purity of the nce is calculated t including such carriers or excipients, To give but one example, in some embodiments, a biological polymer such as a polypeptide or polynucleotide that occurs in nature is ered to be ted" when: a) by virtue of its origin or source of derivation is not associated with some or all of the components that accompany it in its native state in nature; b) it is substantially free of other polypeptides or nucleic acids of the same species from the species that produces it in nature; or c) is expressed by or is otherwise in association with components from a cell or other expression system that is not of the s that produces it in nature. Thus, for instance, in some embodiments, a polypeptide that is chemically synthesized or is synthesized in a cellular system different from that which produces it in nature is ered to be an "isolated" polypeptide, Alternatively or additionally, in some embodiments, a polypeptide that has been subjected to one or more purification techniques may be considered to be an ted" polypeptide to the extent that it has been separated from other components: a) with which it is associated in nature; and/or h) with which it was ated when initially produced. £9998; ”Locus” or "Loci” includes a specific location(s) of a gene (or significant sequence), DNA sequence, polypeptide—encoding sequence, or position on a chromosome ofthe genome of an organism. For example, a "CQORF72 locus" may refer to the specific on of a C90RF72 gene, (39012572 DNA sequence, ’Zn encoding sequen ‘e, or C9ORF72 position on a chromosome of the genome of an organism that has been identified as to where such a sequence resides. A CQQRFW locus may comprise a regulatoiy element of a CQORFH gene, including, but not limited to, an enhancer, a promoter, 5' and/or 3' UTR, or a combination thereof. Those of ordinary skill in the art will appreciate that chromosomes may, in some embodiments, contain hundreds or even thousands of genes and demonstrate physical co—localization of similar genetic loci when comparing between different species. Such genetic loci can be described as having shared synteny. £9999; ”Nonuhumcn animai" includes any vertebrate organism that is not a human.
In some embodiments, a nonuhuinan animal is a cyciostome, a bony fish, a cartilaginous fish (cg, a shark or a ray), an amphibian, a reptile, a mammal, and a bird, in some embodiments, a non—human mammal is a primate, a goat, a sheep, a pig, a dog, a cow, or a rodent. In some embodiments, a nonuhuinan animal is a rodent such as a rat or a mouse. [hillflfll “Nucleic acid "’ includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain. in some embodiments, a "nucleic acid" is a compound and/or substance that is or can be incorporated into an oligonucieotide chain Via a phosphodiester linkage. As will be clear from context, in some embodiments, "nucleic acid" refers to individual nucleic acid residues (egt, nucleotides and/or nucleosides); in some embodiments, "nucleic acid" refers to an oiigonucleotide chain comprising individual c acid residues. in some embodiments, a "nucleic acid" is or ses RNA; in some embodiments, a "trader‘s acid" is or comprises DNA, in some embodiments, a "nucleic acid" is, comprises, or consists of one or more natural c acid residues. In some embodiments, a "nztcl’eic acid" is, comprises, or consists of one or more nucleic acid s, in some embodiments, a nucleic acid analog differs from a /tic acid" in that it does not utilize a phosphodiester backbone. For example, in some embodiments, a ic acid" is, comprises, or consists of one or more impose nucleic acids", which are known in the art and have peptide bonds instead of phosphodiester bonds in the ne, are considered within the scope of the present invention Alternatively or additionally, in some embodiments, a "nucleic acid" has one or more phosphorothioate and/or 5'—N—phosphoramidite linkages rather than phosphodiester bonds. in some embodiments, a "nucleic acid" is, comprises, or consists of one or more l nucleosides {e,g., adenosine, thymidine, ine, cytidine, uridine, deoxyadenosine, deoxythymidine, deoxyguunosine, and deoxycytidine). in some embodiments, a "nucleic acid" is, ses, or consists of one or more nucleoside analogs {e.g., 2—aminoadenosine, 2—thiothymidine, inosine, pyrrolo—pwrimidine, 3mmethyl adenosine, 5~methylcytidine, {3—5 propynyl—cytidine, CnS propynylmuridine, 2n aminoadenosine, CSubroniouridine, C5 "fluorouridine, CS—iodouridine, C5~propynyl~ uridine, C5 —prop}nyl~cytidine, C5 —methylcytidine, 2—aminoadenosine, 7—deazaadenosine, 7-deazaguanosine, Smoxoadenosine, uanosine, O(6)~methylguanine, 2— thiocytidine, methylated bases, intercalated bases, and ations thereof). In some embodiments, a "nucleic acid" comprises one or more modified sugars (eg, 2‘~ fluororibcse, ribose, Z'udeoxyribcse, arahinose, and hexose) as compared with those in natural nucleic acids. in some embodiments, a ”nucleic acid” has a nucleotide ce that encodes a functional gene product such as an RNA or protein, in some embodiments, a "nucleic acid" includes one or more introns. ln some embodiments, a "nucleic acid" includes one or more exons. in some ments, a "rzucleic acid" is prepared by one or more of isolation from a natural source, enzymatic synthesis by polymerization based on a complementary template (in vivo or in , reproduction in a inant cell or system, and chemical synthesis. in some ments, a "nucleic acid" is at least 3, 4, 5, 6, 7, 8, 9, l0, l5, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, ‘75, 00, 85, 90, 95, 100, 110, l20, l30, 140, l50, 160, l70, 180, l90, 20, .225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 600, 700, 800, 900, l000, l500, 2000, 2500, 3000, 3500, 4000, 4500, 5000 or more residues long. in some embodiments, a eic acid" is single stranded; in some embodiments, a ”nucleic acid” is double stranded. in some embodiments, a "nucleic acid" has a nucleotide sequence sing at least one element that encodes, or is the complement of a sequence that encodes, a polypeptide. in some embodiments, a ”nucleic acid” has enzymatic activity. {0010i} "Qpcmbly linked" includes a osition wherein the components described are in a relationship permitting them to function in their intended manner. A control ce "operably links " to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences, "Opel/obit} linked" sequences include both expression l sequences that are contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest. The term "expression control sequence" includes polynucleotide sequences, which are necessary to affect the expression and processing of coding sequen res to which they are ligated. "Expression control sequences" include: riate transcription initiation, termination, er and enhancer ces; ent RNA processing signals such as splicing and polyadenylation signals; sequences that ize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozalt consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion, 'l‘he nature of such control sequences differs depending upon the host organism. For example, in prokaryotes, such control sequen‘es generally e promoter, ribosomal binding site and transcription ation sequence, while in eukaryotes typically such control sequences include promoters and transcription termination sequence. The term "cwztml sequences" is intended to include components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences. {39182} "Phenotype” includes a trait, or to a class or set of traits displayed by a cell or organism. in some embodiments, a particular phenotype may correlate with a ular allele or genotype. ln some emhodiments, a phenotype may he discrete; in some embodiments, a phenotype may be continuous.
Etillllldl "Physiological conditions " es its art—understood meaning reterencing conditions under which cells or organisms live and/or reproduce. in some embodiments, the term includes conditions of the external or internal mileu that may occur in nature for an organism or cell system. in some embodiments, logical conditions are those conditions present within the body of a human or non—human animal, especially those conditions present at and/or within a surgical site. Physiological conditions typically include, e.g, a temperature range ofle 40°C, heric pressure of l, pH of 6 8, glucose concentration of l — 20 rnlvl, oxygen concentration at heric levels, and gravity as it is tered on earth. in some embodiments, conditions in a laboratory are manipulated and/or maintained at physiologic conditions. In some embodiments, logical conditions are tered in an organism. ldtlllldl “Polypeptide" es any polymeric chain of amino acids. ln some embodiments, a ptide has an amino acid sequence that occurs in nature. in some embodiments, a polypeptide has an amino acid ce that does not occur in nature. ln some embodiments, a polypeptide has an amino acid sequence that contains portions that occur in nature tely from one another tie, from two or more different organisms, for example, human and nonuhuman portions). in some embodiments, a polypeptide has an amino acid sequence that is engineered in that it is designed and/or produced through action of the hand of man. llltlltlfil ”Prevent” or fiercest/trims " in connection with the occurrence of a disease, disorder, and/or condition, includes reducing the risk of developing the disease, disorder and/or condition and/or to delaying onset of one or more characteristics or symptoms of the disease, er or condition. Prevention may he considered complete when onset of a disease, er or condition has been delayed for a predefined period oftiine. llltlltldl ”Reference" includes a standard or control agent, animal, cohort, individual, population, sample, sequence or value against which an agent, animal, cohort, individual, population, , sequence or value of st is compared. in some embodiments, a reference agent, animal, cohort, individual, tion, sample, sequence or value is tested and/or determined substantially simultaneously with the testing or determination of the agent, animal, cohort, individual, population, sample, sequence or value of interest. in some embodiments, a reference agent, animal, cohort, individual, population, sample, sequence or value is a historical reference, optionally embodied in a tangible medium. in some embodiments, a reference may refer to a control. A "reference" also includes a "reference animal". A "reference animal" may have a modification as described herein, a modification that is different as described herein or no modification tie, a wild type animal). Typically, as would be understood by those skilled in the art, a nce agent, animal, cohort, dual, population, sample, sequence or value is determined or characterized under conditions able to those utilized to ine or characterize the agent, animal (e.g., a ), cohort, individual, population, sample, ce or value of interest. {@9197} nse" includes any beneficial tion in a subject's ion that occurs as a result of or correlates with treatment. Such alteration may include stabilization of the condition (e.g., prevention of deterioration that would have taken place in the absence of the treatment), amelioration of symptoms of the condition, and/or improvement in the prospects for cure of the condition, etc. it may refer to a subj ect’s response or to a neuronls response. Neuron or subject response may be measured ing to a wide y of criteria, including clinical ia and objective criteria.
Examination of the motor system of a suhject may include examination of one or more of strength, tendon reflexes, superficial reflexes, muscle built, coordination, muscle tone, abnormal movements, station and gait. Techniques for assessing response include, but are not limited to, clinical examination, stretch flex (myotatic reflex), l-loffmann's reflex, and/or pressure tests. Methods and guidelines for assessing response to treatment are discussed in Brodal, A.: Neurological Anatomy in Relation to Clinical Medicine, ed. 2, New York, ()xiord University Press, l969; Medical Council of the UK; Aids to the Examination of the eral Nervous System, Palo Alto, Calif, Pendragon House, 1978; Monradnlérohn, G.H., Refsum, 8.: 'l‘he Clinical Examination of the s System, ed. l2, London, ills; Lewis .81; Co, l964; and Wolf, Eli: Segmental Neurology, A Guide to the Examination and lnterpretation of Sensory and Motor Function, ore, University Park Press, l98l. The exact response criteria can be selected in any appropriate manner, provided that when comparing groups of neurons and/or patients, the groups to be compared are assessed based on the same or comparable criteria for determining response rate. ()ne of ordinary skill in the art will be able to select appropriate criteria. {aortas} “Risk", as will be understood from context, of a disease, disorder, and/or ion comprises likelihood that a particular individual will develop a disease, disorder, and/or condition (e.g., a radiation injury). in some embodiments, risk is expressed as a percentage. in some embodiments, rislt is from 0, l, '2, 3, 4, 5, 6, 7, 8, 9, , 20, 30, 40, 50, 60, ‘70, St), 90 and up to 100%, in some embodiments, risk is expressed as a risk relative to a risk associated with a reference sample or group of reference samples. in some embodiments, a reference sample or group of nce samples have a known risk. of a disease, disorder, condition and/or event (cg, a radiation injury), in some embodiments a reference sample or group of reference samples are trom individuals comparable to a particular individual. ln some embodiments, relative risk is 0,1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. {$6189} "Substantially" includes the qualitative condition of exhibiting total or nearm total extent or degree ofa characteristic or ty of interest. One of ordinary skill in the biological arts will tand that ical and chemical ena rarely, if ever, go to completion and/or proceed to completeness or e or avoid an te result, The term "satiationtrail};n is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena. ltllllllll "Substantial homology" includes a comparison between amino acid or c acid sequences, As will be appreciated by those of ordinary skill in the art, two sequences are generally considered to he "substantialb’ homologous" if they contain homologous es in corresponding positions. Homologous residues may be identical residues, Alternatively, homologous residues may be non ~identical residues with appropriately similar structural and/or onal teristics. For example, as is well known by those of ordinary shill in the art, ‘ertain amino acids are typically classified as "hydrophobic" or "hydrophilic" amino acids, and/or as having " or ”nonwpolur" side chains. Substitution of one amino acid for another of the same type may often be eensidered a ogous" substitutien. Typical amino acid, eategorizatlons are summarized below.
Alanine Ala A Nenpelar Neutral l .8 ne Arg R Felar Positive ~45 Asparagirre Asn N Polar Neutral ~35 Aspartie acid Asp D Pelar Negative ~35 Cysteine Cys C. Neripelar Neutral 2.5 Glutantie acid Gin F Polar Negative “3,5 Glutaniine Gin Q Polar Neutral ~35 Glycine G13/ C ar Neutral ~04 Histidine His ll Felar ye ~32 lsoleueine lie 1 Nonpelar Neutral 45 Leueine Len L N’enpelar Neutral 3.8 Lysine Lys K Polar ve ~39 Methionine Met lVl Nonpolar Neutral l ,9 Phenylalanine Plie F Nonpolar Neutral 2.8 Preline Pre P Nenpelar Neutral —l .6 Serine Set S Felar Neutral ~08 ’l’lireenine 'l‘lir '1‘ Polar Neutral ~(lt7 Tryptephan Trp W N’enpelar Neutral ~ll.9 Tyresine Tyr Y Pelar Neutral —l .3 Valine Val 'V Nonpolar Neutral 4,2 Asparagine er ie aeiri Asa, B Glutarnine er glutalnie acid (Six Z Leueine er lsoleueine Xle J Unspecified or unknown amine aeid Xaa X [llllllll As is well knewn in this art, amino acid er nucleic acid ces may be eempared using any ef a variety (if algeritllms, including these available in commercial computer pregrarns such as BLASTN for tide sequences and BLASTF, gripped BLAST, and FSl~BLAST for amino acid sequences. Exemplary such programs are described in ul, S. F. et al., 1990, .1. Mol. Biol, 215(3): 403-410; Altschul, S. F. et al., 1997, s in Enzymology; Altschul, S. F. et a1, 1997, Nucleic Acids Res, 9—3402; Baxevanis, All, and B. F. F. Guellette (eds.) forn1atics: A Practical Guide to the Analysis of Genes and Proteins, Wiley, 1998; and Misener et al. (eds) Bioini‘ormatics Methods and Protocols ds in Molecular Biology, Vol. 132), l-lurnana Press, 1998. In addition to identifying homologous sequences, the programs mentioned above typically provide an indication of the degree ofhomology. in some embodiments, two ces are considered to be substantially homologous if at least 50%, 55%, 60%, 65%, 70%, 75%, 889’, 35%, 981%, 91%, 92%, 93%, 94%, 959’, 96%, 97%, 98%, 99% or more of their ponding residues are homologous over a relevant h of residues. in some embodiments, the relevant stretch is a complete ce. in some embodiments, the relevant stretch is at least 9, 10, ll, 12, 13, l4, 15, 16, l7 or more residues. ln some embodiments, the relevant stretch includes contiguous residues along a complete sequence. in some embodiments, the nt stretch includes discontinuous residues along a complete sequence, for example, noncontiguous residues brought together by the folded conformation Of a polypeptide or a portion thereof. in some embodiments, the relevant stretch is at least 10, 15, 2t), 25, 30, 35, 40, 45, fill, or more residues. {@9112} "Substantial ?" includes a comparison between amino acid or nucleic acid sequences. As will be appreciated by those of ordinary skill in the art, two " if they contain sequences are generally considered to be antially identical identical residues in corresponding ons. As is well known in this art, amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those ble in commercial computer programs such as BLASTN for nucleotide sequences and BLAS’l‘l’, gapped BLAST, and Fill—BLAST for amino acid sequences. ary such programs are described in Altschul, 5.17. et al., 1990, .l. Mol. Biol., 2l 5(3): 463—410; Altschul, SF. et al., 1997, Methods in Enzymology; Altschul, SF. et al., l997, Nucleic Acids Res, 25:3389—3402; Baxevanis, All, and Bill”. Ouellette (eds) Bioinformatics: A Practical Guide to the Analysis of Genes and Proteins, Wiley, 1998; and Misener et al. (eds) Bioinforrnatics Methods and Protocols {Methods in Molecular Biology, Vol. 132), Humana Press, 1998. ln addition to identifying identical sequences, the programs mentioned above typically provide an indication of the degree of identity. In some embodiments, two sequences are considered to be substantially identical if at least 50%, 550/, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of their corresponding residues are identical over a relevant stretch of residues. in some embodiments, the relevant h is a te ce. in some emhodiments, the relevant stretch is at least l0, l5, 20, 25, , 35, 40, 45, 50, or more residues. {can 3} ”Targeting vector" or "targeting cortstmcr" includes a pol ynucleotide molecule that comprises a targeting region, A targeting region comprises a sequence that is identical or substantially identical to a sequence in a target cell, tissue or animal and provides for integration of the targeting construct into a position within the genome of the cell, tissue or animal via homologous recombination. Targeting regions that target using sitenspecitic recombinase recognition sites (cg, lax? or Frt sites) are also ed. In some embodiments, a targeting construct as described herein further comprises a nucleic acid sequence or gene (eg, a reporter gene or homologous or logous gene) of particular interest, a selectable marker, control and or regulatory sequences, and other nucleic acid sequences that encodes a inase or recombinogenic protein. in some embodiments, a targeting construct may comprise a gene of interest in whole or in part, wherein the gene of st encodes a ptide, in whole or in part, that has a similar function as a protein encoded by an endogenous sequence. In some embodiments, a targeting construct may comprises a humanized gene of interest, in whole or in part, wherein the zed gene of interest encodes a polypeptide, in whole or in part, that has a r firnction as a polypeptide encoded by an endogenous sequence. in some embodiments, a targeting construct may comprise a reporter gene, in whole or in part, wherein the reporter gene encodes a ptide that is easily fied and/or measured using techniques known in the art. {$6114} genic entrant", ”transgenic non—human animal” or "13+" includes any nonunaturally occurring nonuhuman animal in which one or more of the cells of the nonhuman animal contain heterologous nucleic acid and/or gene encoding a polypeptide of interest, in whole or in part, in some embodiments, a heterologous nucleic acid and/or gene is introduced into the cell, directly or indirectly by introduction into a precursor cell, by way of deliberate c manipulation, such as by microinjection or by infection with a recombinant Virus The term genetic manipulation does not include classic breeding techniques, but rather is directed to introduction of inant DNA molecule(s). This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA, The term "212+" includes animals that are zygous or homozygous for a heterologous nucleic acid and/or gene, and/or animals that have single or multi—copies of a heterologous nucleic acid and/or gene. {$6115} "Treatment”, " or "Treating" includes any administration of a substance ( e.g, a therapeutic candidate) that partially or completely alleviates, ameliorates, s, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a. particular disease, disorder, and/or condition. ln some embodiments, such treatment may be administered to a subject who does not exhibit signs of the relevant disease, disorder and/or condition and/or of a subject who exhibits only early signs of the e, disorder, and/or condition. Alternatively or additionally, in some embodiments, treatment may be administered to a t who ts one or more established signs of the relevant disease, disorder and/or condition. in some embodiments, treatment may be of a t who has been diagnosed as suffering from the nt disease, disorder, and/or condition, in some embodiments, treatment may be of a subject known to have one or more tibility factors that are statistically correlated with increased risk of pment of the relevant disease, disorder, and/or condition. {$6116} "Variant” includes an entity that shows significant structural identity with a reference entity, but differs urally from the reference entity in the presence or level of one or more chemical moieties as compared with the reference entity. In many embodiments, a. "variant" also differs lunctionally from its reference entity. in l, whether a particular entity is properly considered to he a "variant" of a reference entity is based on its degree of structural identity with the reference entity. As will be appreciated by those skilled in the art, any biological or chemical reference entity has certain characteristic structural elements. A "variant", by tion, is a ct chemical entity that shares one or more such characteristic structural elements. To give but a few examples, a small le may have a characteristic core structural element (e.g., a rnacrocycle core) and/or one or more characteristic pendent moieties so that a variant of the small molecule is one that shares the core ural element and the characteristic pendent moieties but differs in other pendent moieties and/or in types ofhonds present {single vs. double, E vs, Z, etc.) within the core, a polypeptide may have a characteristic sequence element comprised of a plurality of amino acids having designated positions relative to one another in linear or three—dimensional space and/or contributing to a particular biological on, a nucleic acid may have a characteristic sequence element comprised ofa plurality of nucleotide residues having designated positions relative to on another in linear or threevdimensional space. For example, a mpolypeptide" may differ from a reference polypeptide as a result of one or more differences in amino acid sequence and/or one or more differences in al moieties (cg, carbohydrates, lipids, etc.) covalently attached to the polypeptide backbone. In some embodiments, a "variant polypeptide" shows an overall sequence ty with a. reference polypeptide that is at least 85%, 86%, 8 %, 88%, 890/, 96%, 9l%, 92%, 93%, 9 %, 95%, 969/, 97%, or 99%. Alternatively or additionally, in some embodiments, a "variant ptide" does not share at least one characteristic sequence element with a reference polypeptide. in some embodiments, the reference ptide has one or more biological activities. in some embodiments, a "variant polypeptide" shares one or more of the biological activities of the reference polypeptide. in some embodiments, a "variant pub/peptide" lacks one or more of the biological activities of the reference polypeptide. ln some embodiments, a nt polypeptide" shows a reduced level of one or more biological ties as compared with the referenie polypeptide. in many embodiments, a polypeptide of interest is considered to be a "variant" of a parent or reference polypeptide if the polypeptide of interest has an amino acid sequence that is identical to that of the parent but for a small number of sequence alterations at particular positions.
Typically, fewer than 20%, 15%, “3%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, or 2% of the residues in the variant are substituted as compared with the parent. in some embodiments, a nt" has ll), 9, 8, 7, 6, 5, 4, 3, 2, or l substituted residue(s) as compared with a parent. fiften, a "variant" has a very small number (cg, fewer than 5, 4, 3, 2, or l) number of substituted functional residues (i.e., residues that participate in a particular ical activity), Furthermore, a nt" typically has not more than 5, 4, 3, 2, or l additions or ons, and often has no ons or deletions, as compared with the parent. Moreover, any additions or deletions are typically fewer than about 25, about 20, about l9, about l8, about 17', about l6, about l5, about l4, about l3, about it), about 9, about 8, about 7, about 6, and commonly are fewer than about 5, about 4, about 3, or about 2 residues. in some embodiments, the parent or reference polypeptide is one found in nature, As will be understood by those of ordinary skill in the art, a plurality of variants of a particular ptide of interest may commonly be found in nature, particularly when the polypeptide of interest is an infectious agent polypeptide. liltill’l’} r" includes a nucleic acid molecule capable of transporting another nucleic acid to which it is associated. in some embodiment, vectors are capable of extra~ chromosomal replication and/or expression of nucleic acids to which they are linked in a host cell such as a eukaryotic and/or proliaryotic cell. Vectors capable of directing the expression of operahly linked genes are referred to herein as "expression vectors." [llllll 8} "Wild type" includes an entity having a structure arid/or activity as found in nature in a "normal" (as contrasted with mutant, diseased, altered, etc.) state or t.
Those of ordinary shill in the art will appreciate that wild type genes and polypeptides ot‘ten exist in multiple different forms (cg, alleles).
DETAILED DESCRIPTIGN 0F CERTAEN Eh/lBOD‘lh/lENTS {@9119} Non-human animals are provided having a disruption in a C9GRF72 locus. ln particular, non—human animals described herein have a deletion of an entire coding sequence in a C90RF172 locus, i.e., a deletion of a genomic segment coding for all C90RF172 isoforrns (e.g., a deletion of the coding portion of exon 2 through the coding portion of exon ll of isoforni Vl). Non—human animals as described herein demonstrate weight loss and ALE—like motor alites such as, for example, motor inactivity and gait impairment. Also, non-human animals described herein demonstrate megaly and/or lymphadenopathy at about eight (8) weeks of age. Further, nonhuman animals described herein trate glornerulonephiitis at about 35 weeks of age. Therefore, provided non—human animals are particularly usetul for the development and identification of therapeutic candidates for the treatment and/or amelioration of neurodegenerative diseases, disorders and conditions and, in some embodiments, autoimmune and/or atory diseases, disorders and ions. in particular, non— human s described herein encompasses the introduction of a reporter gene into an endogenous CQORF72 locus ing in expression of the reporter gene tie, a reporter ptide) in the nervous and e systems of the non—human animal. Such transgenic nonuhurnan animals provide a source of cells for ining the efficacy of therapeutic candidates to ameliorate ALS arid/or FTD. Further, such transgenic non— human animals provide a useful animal model system for the pment of therapeutics for the treatment of egenerative, autoimmune and/or matory diseases, disorders and conditions. fiddlZlB} in some embodiments, non—human animals bed herein develop ALSH and/or ETD—like disease due to the absence of, lacl: of, or decreased level ol‘CQtlkFVZZ (cg, RNA, polypeptide, etc.) in cells of the non—human animals. in some embodiments, man animals described herein develop glomerulonephritis due to the absence of, lack of, or se level oi‘C90RF72 {e.g., RNA, polypeptide, etc}. in some embodiments, non—human animals described herein are heterozygous for a disruption in a CQORF72 locus as described herein. in some embodiments, non—human animals described herein are homozygous for a disruption in a C9QRF72 locus as described herein. In some embodiments, nonuhuman animals as described herein comprise a er gene, in whole or in part, wherein said reporter gene is operably linked to a (39013572 promoter. in some embodiments, C9GRF72 promoters include endogenous C90RF72 promoters. [littlzll The present disclosure provides a hensive phenotypic analysis of a non—human animal line with global CQQWZ ablation and the discovery of a unique role for C9mj72 in innnune system homeostasis. The present disclosure specifically trates that a complete ablation of C90iff72 resulted in gait abnormalities and an indication towards hindlimh weakness that suggests possible onset of lower motor neuron pathology beginning at about 40 weeks of age. As described herein, the immune ype exhibited by C9w3f72'” nonuhuman animals ted of select expansions in both myeloid and lymphoid compartments, with increased activation of T cells and ed plasma cells. Neutrophilia and monocytosis resulted in mixed infiltrates in immune organs that expanded, but did, not obliterate basic architecture. C90ijf72'/' non— human animals demonstrated elevated serum cytokines (cg, lit—l2) and tissue RNA signatures consistent with myeloid upregulation. Renal disease with accompanying pathological changes such as, for example, ned basement membrane and cast formation, was t in the majority ol’nonuhuman animals by about 35 W66l§S of age.
At a copic level, glomeruli stained heavily with lgG and light antibody and complement C3 in a pattern indicating tion of immune complexes. Al so described herein, C905f72'/" non—human animals demonstrated, high titer of autoantibodies including anti~RF, ANA, anti—Sm, and anti—cardiolipin, which indicated that loss of 72 expression profoundly disrupts immune tasis. None of the immune~ related phenotype characteristics were observed in wild type or 2+fl (heterozygous) non—human animals. Thus, the present ion provides, among other thingsg the creation of an improved in viva system for the development ofnew ies and/or fication ofnew eutic targets for the treatment of various diseases, disorders or ions that are not achievable h the use of established in viva systems due? in part? to the nonexistence ofparticular phenotypes in such established systems. {@9122} Various aspects of the invention are described in detail in the following sections. The use of sections is not meant to limit the invention. Each section can apply to any aspect of the invention. in this applicationg the use of "or" means "and/or" unless stated otherwise.
C993”??? {@9123} Amyotrophic lateral sclerosis (ALS), also referred to as Lou Gehrig's disease? is the most frequent adult~onset paralytic disorder, characterized by the loss of upper and/or lower motor neurons. ALS occurs in as many as 2&000 individuals across the United States with about 59,009 new cases occurring each year. Frontotemporai dementia (ETD), originally referred to as ?ick’s disease after ian Arnold Pick? is a group of disorders caused by progressive cell degeneration in the frontal or temporal lobes of the brain. FTD is reported to count for l0~l 5% of all dementia cases. GGGGCC hexanucleotide repeat expansion between two non—coding exons of CQORFH have been linked to both ALS and ETD (Delesusnllernandez, M. et alj 2011, Neuron 722245—256; , AE et at, Ztll l, Neuron 721257—263; Maiounie, E. et ai., 20l23 Lancet Neurol. l l :323—330; Waite, All et al., 20%, Neurobiol. Aging 35:l779.e5—l779.e13). However, the mechanism through which such repeat mutations cause disease, whether through a loss— or gain—oiliunction oftoxicity, remains unclear. For example, ish with reduced C9ORF'72 expression from targeted reduction or knockdown demonstrate axonopathy and ts in locomotor firnction (Ciura, S» et al.3 2M3? Ann Neurol. 74(42): 180—187} while mice with reduced (3903572 expression do not show any behavioral or pathological es associated with ALS disease r—Tourenne, C. et al., 20%, Proc. Nat. Acad. Sci. USA. E4530—E4539). Further, knock—in mice that have been generated to contain 66 (790RF72 repeat expansions exhibit RNA foci and dipeptide protein ates in their s These mice showed cortical neuron loss and exhibited behavior and motor deficits at 6 months of age (Chew, J. et ala 2915, Science May l4. Pii:aaa9344)., 1611124} Many pathological aspects related to GGGGCC hexanucleotide repeat expansions in (3.901113772 have been reported such as, for example, repeat length— dependent formation of RNA foci, sequestration of specific RNA—hinding proteins, and accumulation and aggregation of dipeptide repeat proteins (cg, reviewed in Stepto, A. et al., 2014, Acta Neuropathol. —339; see also Almeida, S. et al., 2013, Acta Neuropathol. 126385—399; Bieniek, KP. et a1, 20M, JAMA Neurol. 71(6): 775—78l; van rswijk, M. et al., 21114, Mol. Neurodegen. 9:38, 10 pages). Although C9ORE72 has been reported to regulate endosomal trafficking (Farg, M..A. et al, 2014, Human Mol. Gen. :3579_3595), much of the cellular function oi‘C90RF'72 remains unknown. lndeed, C90RF72 is a gene that s an uncharacterized protein with unknown litnction. Despite the lack of understanding surrounding C9ORF72, l animal models, including engineered cell lines, for ALS and/or FTD have been developed son, Ell, 2612, Ann. Neurol. 72(6):837~849; Panda, SK. et al., 2013, Genetics l95:7(13~715; Suzuki, N. et al, 2013, Nature Neurosci. 16(12): l725~1728g Xu, Z. et al., 2013, l’roc. Nat. Acad. Sci. USA. 110(19):7778—7783; Hukeina, R11. et al., 2014, Acta athol. Comm. 2: 166, 4 pages). For example, transgenic mice having a Zan gene insertion that replaced exons 2—6 of (390672 (3110040211‘ilik) have been produced by gene targeting efforts from the Knockout Mouse Project (see Figure ld of Suzuki, N. et al., 2013, supra; for KGMP, see Skarnes, W1C. et al., 2011, Nature 474(7351):337~342). X—gal ng was observed in the brain, spinal cord, testis and germinal centers of the spleen in these mice, however, not in muscle lis anterior), heart, lung, liver, and kidney. it is unclear, however, if the remaining undeleted exons (i.e., 7—1 1) yie1d any expression and, consequently, function. Another report using a transgenic mouse strain containing 81) GGGGCC repeats operahly linked with a fluorescent reporter and controlled by a tetracycline sive element without any surrounding C9my‘72 sequences demonstrated neuronal cytoplasmic inclusions similar to those seen in ALS~FTD patients, which ts that the repeat expansion itself may be responsible for disease a, RR; et al., 2014, Acta athol. Comm. 2:166, 4 pages). These mice have been useful to establish an initial C90ijf72 expression profile in cells of the CNS and provide some understanding of the mechanism of action associated with the repeat expansion; however, it remains unclear if the specific design of these constructs in these mice indicate specific functions that can he confidently uted to C905f72 or are a result of something else, unrelated to 2 lunction. fitltlflfi} In some cases, however, construct design can inlluence the phenotype of the resulting transgenic animal (see, cg, Muller, ll, 1999, Mech. Develop. 8l .
Because the (7.90;:f72—disrupted mice as described above utilized a targeting vector with a promoter—driven selection cassette (see Figure ld of Suzuki, N et al., 203, supra; Figure 1 of Skarnes, W.C. et al., lel 1, supra), it is not clear if the displayed expression pattern correctly correlates with normal C90RF72 expression. indeed, expression of the remaining exons 7~ll driven by the selection cassette—associated promoter or C90RF72 er itself has not been determined. As a result, phenotypes, and perhaps C90RF72 expression via incl, ed in such mice may be modified or otherwise skewed due to aspects of the targeting vector. Further, a transgenic mouse strain containing an inducible GGGGCC repeat (liulrema, 2614, supra) was designed t human flanking sequen re ably due to the fact that such surrounding sequence was thought to affect translation of repeat sequences. Thus, such in viva s exploiting C90RP72—mediated biology for therapeutic applications are incomplete, fitltllzo} As described herein, the present disclosure specifically describes a non~ human animal model for ALS and/or ETD, which nonuhuman animal comprises a disruption in a C9ORF72 locus, ln particular, the present disclosure specifically describes a rodent model for ALS and/or ETD, wherein the rodent comprises a deletion of the entire coding sequence for all C9riigf72 isoforrns (cg, the coding portion ofexon 2 through the coding portion ofexon ll ofV l) of a 772 gene via insertion of a 25262 er gene, The targeting vector ed by the t inventors was designed to contain a sell‘ldeleting drug selection cassette (see ego, US. Patent Nofs 8,697,851, 8,518,392 and 8,354,389, all ofwhich are herein incorporated by reference), which allows for removal of the thug selection cassette in a pment—dependent manner, thereby removing any chance oter affects or aberrant expression of remaining exons or effects from the ion cassette itself. As described herein, the present inventors achieved complete ablation of ($0572 Further, the t inventors measured the motor behaviors of these rodents using rotarod, open field and CatWalk assays up to 60 weeks of age and neurological deficits throughout the same period.
Without wishing to be bound by any particular theory, the present disclosure demonstrates that only 4 % of C90rjf72 knockout rodents survived past so weeks of age and the s ceased gaining body weight beginning around 40 weeks ot‘age. While rotarod tests did not show significant changes due to C9orff72 deletion, rodents described herein demonstrated significant hind limb paresis, motor impairment, decreased mobility and gait abnormalities at arotmd 50 weeks ofage. Further, the present disclosure specifically demonstrates that genetic ing of murine 2 results in multiple motor and neurological abnormalities similar to those found in human motor neuron es. Thus, rodents described herein e, at least in some embodiments, improved in viva systems for development of therapeutic candidates for the treatment oi” neurodegenerative disease such as, for example, ALS and/or FTD. Further, rodents described herein overcome deficits in existing animal models characterized by imal {790572 deletion for the development ol‘CQortVZutargeted ies. (3903?F72 Sequences {00127} Mouse C9GRF72 transcript variants have been reported in the art (e.g., Koppers et al., Ann Neurol (20l5); 78: 426—438; Atkinson et al,, Acta Neuropathologica Communications (20l5) 3: 59), and are also depicted in Figure lA. The genomic information for the three reported mouse {1901932772 transcript variants is also available at the Ensemhl web site under designations of ENSMUSTOOGOOl 08l27 (Vi), ENSMU8300000 108 l 26 (V2), and ENSMU 5100000084724 (V3). Exemplary non— human (rag, rodent) C9GRF72 mRNA and amino acid sequences are set forth in Table l, For mRNA sequences, hold font contained within parentheses indicates coding ce and utive exons, where indicated, are separated by alternating lower and upper case letters. For amino acid sequences, mature polypeptide sequences, where indicated, are in bold font. {00128} Human C90RF72 ript variants are lrnown in the art. Qne human C90RF72 transcript variant lacks multiple exons in the central and 3’ coding regions, and its 3' al exon extends beyond a splice site that is used in variant 3 ( see below), which results in a novel 3* untranslated region (U’l‘ll) as compared to variant 3. This t encodes a significantly shorter polypeptide and its Cuterminal amino acid is distinct as compared to that which is encoded by two other variants. The mRNz—‘t and amino acid sequences of this variant can be found at k accession numbers NM_l45005,6 and NP_659442.2, tively, and are hereby incorporated by reference. A second human C9QRF72 transcript variant (2) differs in the 5' slated region (UTR) compared to variant 3. The mRNA and amino acid sequences of this variant can be found at GenBank accession numbers NMfil 8325A and NP_060795,l, respectivehg and are hereby inecrporated by reference. A third human C9GRF72 transcript variant (3) contains the icngest sequence among three reported variants and encodes the longer isnfnnn, 'I‘he mRNA, and amine acid ces of this variant can be found at GenBank accession numbers NM___{){)1256054.2 and NP___001242983.1, respectively, and are hereby crated by renerenee. Variants 2 anti 3 encode the same pretein.
TABLE 1 Mus museums C90nf72 rnRNA (NM___G€E£€E81343; SEQ ID Nflzi) gtgtecggggeggggcggtcceggggeggggcecggagcgggctgcggttgeggteectgcgecggcggtgaaggcge agcagcggcgagtggC’I‘A’I’TGCAAGCGTTCGGA’I’AA’I‘G’I‘GAGACC’E‘GGAA’I‘GCAG’I‘G GGGATGCAGGG(ATGTCGACTATCTGCCCCCCACCATCTCCTGCT AAGACAGAGATTGCTTTAAGTGGTGAATCACCCTTGTTGGCGG CTACCTTTGCTTAC’I‘GGGA’E‘AA’E‘ATECTTGGTCCTAGAGTAAGGCATATT TGGGCTCCAAAGACAGACCAAGTGCTTCTCAGTGATGGAGAAATAACTT E‘TGCCAACCACACTC'E‘AAATGGAGAAATTCTTCGAAATGCAGAGAG TGGGGCTATAGATGTAAAATTTTTTGTCTTATCTGAAAAAGGGGTAATTA TTGTT13CA’E‘TAA’E‘C’1‘TC.GACGGAAACTGGAATGGAGA’E‘CGGAGCAC’E"1‘A TGGACTA'i‘CAATTATACTGCCGCAGACAGAGCTGAGCTTCTACC’E‘CCCA CTTCACAGAGTGTGTGTTGACAGGCTAACACACATTATTCGAAAAGGAA GAATA’E‘GGA'E‘GCA'E‘AAGgaaagaeaagaaaatgteeagaanattgtettggaaggnacagagagga‘t ggaagateagfiGTCAGAGTATCATTCCCATGCTTACTGGGGAAGTCATTCCTG ’E‘AATGGAGCTGCTEGCA’E‘CTATGAAA’E‘CCCACAG’E‘G’E"E‘CCTGAAGACAT TGATatagetgataeagtgeteaatgatgatgaeattggtgaeagetgteaegaaggetttettetcaaTGCCAT CAGCTCACACC’1‘GCAGACCTGTGGC'1‘GTTCCGT'E‘G’E‘AGTTGGCAGCAGT GCAGAGAAAGTAAATAAGatagtaaganegetgtgeetttttetgaeaceageagagaggaaa‘tgcte caggetgtgtgaageagaatcgtectttaagtaegantngggactetttgtgeaaggettgetaaagGA’i‘GCAAC AGGCAGTTTTGTCCTACCCTTCCGGCAAGTTATGTATGCCCCG’FACCCC ACCACGCACAT’E‘GATGTGGA’E‘G’E‘CAACAC'E‘G’E‘CAAGCAGATGCCAC(SGT GTCATGAACA’I‘A’E‘ITA’E‘AATCAACGCAGATACA’E‘GAGGTCAGAGCIGAC AGECTTCTGGAGGGCAACTTCAGAAGAGGACATGGCGCAGGACACCAT CATC’E‘ACACAGA’E‘GAGAGC’E‘TCACTCCTGA’E"E‘Tgantatttteeaagatgtcttaeacaga gaeaetetagtgaaageetteetggateagfll‘CTTCCATTTGAAGCCTGGCCTGTCTCTCA GGAGTACTTTCC’1"E‘GCACAG’E"E‘CCTCCTCAT’E‘CTTCACAGAAAAGCCTTG ACACTAATCAAGTACATCGAGGATGATACgcagaaggggaaaaageectttaagtetette ggaaeetgangatagatettgatttaaeageagagggcgatettaaeataataatggetetagetgagaaaattaagce aggeetaeactetttcatetttgggagaeetttetaeaetagtgtacaagaaegtgatgttetaatgaeettttga}cegtgt etgtgtetgtctetteaeagteaeacetgetgttaeagtgtctcageagtgtgtgggeaeatccttceteecgagteetgct gcaggaeagggtacactacacttgteagtagaagtctgtacctgatgtcaggtgeatcgttaeagtgaatgaetctteetagaata gatgtactcttttagggccttatgmacaafiatcctaag’zactattgctgtcttttaaagatatgaatgatggaatatacacttgaccat aactgatgariggmmgttitgttttgtttgtfltattggaaacttatgattcctggfltacatgiacsacactgaaacca‘mgitagctt‘t acagataaagtgtgagi’tgacttcctgcacctctgtgttctgtggtatgtccgattacflctgccacagctaaacattagagcatttaa agtttgcagttcctcag321aggaaettagtctgactacagafiagficfigagagaagacactgatagggcagagetg‘raggtga aatcagttgttagccam:ctttatagacgtagtccttcagattcggtctgtacagaaatgccgaggggtcatgcatgggacctgag gacetgtgacaagtfimgfiggmafigtagfictgtcaaagaaagtggcamg‘tmtatmfigigccaactmaaggfi aaimcattamttgagccgaattaaaatgcgfi:acctcctgtgmittcccaatcttggaaaatataaificttggcagagggtcaga Ettaagggcccagtcacfitcatctgaccacccfitgcacggatgccgtgtgcctggcfiagafiagaagtccttgttaagtatgtca gagtacaflcge‘tgataagatcti‘tgaagagcagggaage:gcfigactcfitcafitggflmtgcatg’iactctggtgflmccgtgfi cacctgcatcataggaacagcagagaaatctgacc-cagtgctafitttctaggtgctactatggcaaactcaagfggtctgtttctgt 'Ecctgtaacgttcgactatctcgctaga‘tgtgaagtac'Egaflag:ggagttatgtgsaacagcagtgfiaggagtatacacaaaaac aaatatgtgmctatttaaaactgtggacitagcataaaaagggagaatatatttafittfiacaaaagggataaaaatgggcmagi; tectcaccsaceagafiiagcgagaaaaagcmctafietgaaaggtcacgg‘:ggctttggcattacaaatcagaacaacacaca Cigaccatgatggcfigtgaaetaactgcaaggcactccgtcatggtaagcgagtaggtcccacctactagtgtgccgctcattg emacacag‘ragaatcttafiigagtgctaa’ctgfigtcmgctgcmaetgtgttgfiatagaaaatgtaagctgtacagtgaataag 'Eta‘ttgaagcatgtgtaaacactgflatata‘tcm‘mtcctaga‘tggggaaflttgaataaaatacctttgaaai‘tg:tgtgt Mus us (figuri’iz amimp amid {NMfim {331343; SEQ ID N0:2) MSTICPFPSPAVAK’E‘EHALSGESFLLAATFAY‘WDNHJGPRVRHE‘WAPKTDQV LLSDGEITFLANHTLNGEILRNAESGAIDVKFFVLSEKGVHVSLEFDGN‘WNG DRS'E‘E’GLSHLPQTELSFYLPLHRVC‘VDRLTHIHRKGRE‘WMHKERQENVQKH VLEGTERNIEDQGQSH?EWLTGE§W?VNIELLASMKSHSVPEDEDEABTVLNDD BIGBSCHEGELLNAISSHLQTCGCSVVVGSSAEKVNKIVR’E‘LCLFLTPAERK CSRLCEAESSFKYESGLFVQGLLKDATGSFVLFFRQVB/IYAPYPTTHIDVDVN ’E‘VKQMPPCHEHIYNQRRYR’ERSELTAFW'RATSEEDNEAQDTHYTDESFTPDL LHRE}TLVKAFLDQVFHLKPGLSLRSTFLAQFLLILHRKALTLEKYIE DD’E‘QKGKKPEKSLRNLKIDLDLTAEGDLNHMALAEKIKPGLHSFEFGRPFY’E‘SVQ ERDVLMTF Ratms narvegicus (39:05??? mRNA (NMMMBWM'J’GZ; SEQ ED N033) TAGTGTCAGCCATCCCAATTGCCTGTTCCTTCTCTGTGGGAGTOGTG TCTAGACAG’I‘CCAGGCAGGGTA’I‘GCTAGGCAGGTGCGT1"I"1‘GGTTGCC'I’CAG A'I‘CGCAAC’I‘TGAC'I'CCA'R‘AACGGTGACCAAAGACAAAAGAAGGAAACCAGA TTAJMXAAGAACCGGACACAGACCCCTGCAGAATCTGGAGCGGCCGTGGTTG GGGGCGGGGCTACGACGGGGCGGACTCGGGGGCG’I‘GGGAGGGCGGGGCCG GGGCGGGGCCCGGAGCCGGCTGCGGTTGCGGTCCCTGCGCCG(S'CGGTGAAG GCGCAGCGGCGGCGAGTGGCTA’I‘TGCAAGCGT'1’1‘6GATAA’I’GTGAGACCTGG GATGCAGGG(ATGTCGACTATCTGCCCCCCACCATCTCCTGCTGTTGCCA AGACAGAGATTGCTTTAAGTGGTGAATCACCCTTGTTGGCGGCTACCTT TGCTE‘ACTGGGATAA’I‘A'E"E‘C’FE‘GG'E‘CCTAGAGTAAGGCACA’ETTGGGCT CCAAAGACAGACCAAGTACTCCTCAGTGATGGAGAAATCACTTTTCTTG CCAACCACACTCTGAA’E‘GGAGAAAT’E‘CT'1‘CGGAA’E‘GCGGAGAGTGGGGC TGTAAAGTTTTTTGTCTTATCTGAAAAGGGCGTCATTATTGTTT CATTAATCTTCGACGGGAACTGGAACGGAGATCGGAGCACTTACGGACT ATCAATTATACTGCCGCAGACGGAGCTGAGTTTCTACCTCCCACTGCAC AGAGTGTGTGTTGACAGGCTAACGCACATCATTCGAAAAGGAAGGATAT GGATGCACAAGGAAAGACAAGAAAATGTCCAGAAAATTGTCTTGGAAGG CACCGAGAGGATGGAAGATCAGGGTCAGAGTATCATCCCTATGCTTACT GGGGAGGTCATCCCTGTGATGGAGCTGCTTGCGTCTATGAGATCACACA GTGTTCCTGAAGACCTCGATATAGCTGATACAGTACTCAATGATGATGA CATTGGTGACAGCTGTCATGAAGGCTTTCTTCTCAATGCCATCAGCTCA CATCTGCAGACCTGCGGETGTTCTGTGGTGGTAGGCAGCAGTGCAGAGA ATAAGATAGTAAGAACACTGTGCCTTTTTCTGACACCAGCAGA GAGGAAGTGCTCCAGGCTGTGTGAAGCCGAATCGTCCTTTAAATACGAA TCTGGACTCTTTGTACAAGGCTTGCTAAAGGATGCGAflTGGCAGTTTTG TACTACCTTTCCGGCAAGTTATGTATGCCCCTTATCCCACCACACACATC GATGTGGATGTCAACACTGTCAAGCAGATGCCACCGTGTCATGAACATA ATCAACGCAGATACATGAGGTCAGAGCTGACAGCCTTCTGGAG GGCAACTTCAGAAGAGGACATGGETCAGGACACCATCATCTACACAGAT GAGAGCTTCACTCCTGATTTGAATATTTTCCAAGATGTCTTACACAGAGA CACTCTAGTGAAAGCCTTTCTGGATCAGGTCTTCCATTTGAAGCCTGGC CTGTCTCTCAGGAGTACTTTCCTTGCACAGTTCCTCCTCATTCTTCACAG AAAAGCCTTGACACTAATCAAGTACATAGAGGATGACACGCAGAAGGGG AAAAAGCCCTTTAAGTCTCTTCGGAACCTGAAGATAGATCTTGATTTAAC AGCAGAGGGCGACCTTAACATAATAATGGCTCTAGCTGAGAAAATTAAG CCAGGCCTACACTCTTTCATCTTCGGGAGACCTTTCTACACTAGTGTCCA AGAACGTGATGTTCTAATGACTTTTTAAyMLMKYKKfljTGCTCCGTGTGTC TCATGACAGTCACAETTGCTGTTACAGTGTCTCAGCGCTTTGGACACATCCTT CCTCCAGGGTCCTGCCGCAGGACACGTTACACTACACTTGTCAGTAGAGGTC TGTACCAGATGTCAGGTACATCGTTGTAGTGAATGTCTCTTTTCCTAGACTAG ATGTACCCTCGTAGGGACTTATGTTTACAACCCTCCTAAGTACTAGTGCTGTC TTGTAAGGATACGAATGAAGGGATGTAAACTTCACCACAACTGCTGGTTGGT TTTGTTGTTTTTGTTTTTTGAAACTTATAATTCATGGTTTACATGCATCACACT GAAACCCTAGTTAGCTTTTTACAGGTAAGCTGTGAGTTGACTGCCTGTCCCTG TGTTCTCTGGCCTGTACGATCTGTGGCGTGTAGGATCACTTTTGCAACAACTA AAAACTAAAfiCACTTTGTTTGCAGTTCTACAGAAAGCAACTTAGTCTGTCTG CAGATTCGTTTTTGAAAGAAGACATGAGAAAGCGGAGTTTTAGGTGAAGTCA GGATCTTCCTTTATAGACTTAGTCCTTTAGATGTGGTCTGTATAGAC ATGCCCAACCATCATGCATGGGCACTGAATATCGTGAACTGTGGTATGCTTF TTGTTGGTTTATTGTACTTCTGTCAAAGAAAGTGGCATTGGTTTTTATAATTG TTGCCAAGTTTTAAGGTTAATTTTCATTATTTTTGAGCCAAATTAAAATGTGC ACCTCCTGTGCCTTTCCCAATCTTGGAAAATATAAJTTCTTGGCAGAAGGTCA GATTTCAGGGCCCAGTCACTTTC§TCTGACTTCCCTTTGCACAGTCCGCCATG GGCCTGGCTTAGAAGTTCTTGTAAACTATGCCAGAGAGTACATTCGCTGATA AAATCTTCTTTGCAGAfiKMfiKhMlAGCTTCTTGCCTCTTTCCTTTCATTTCTGC CTGGAC'i‘TTGGTG’i‘TCTCCACGTTCCCTGCATCC'I’AAGGACAGCAGGAGAAC TCTGACCCCAGTGCTATTTCTCTAGGTGCTATTGTGGCAAACTCAAGCGGTCC GTCTCTGTCCCTG’TJMXCGTTCGTACCTTGCTGGCTGTGAAGTACTGACTGGTA AAGC'l‘CCG'I‘GC'l'ACAGCAGTGTAGGGTA’I'ACACAAACACAAGTAAG’l‘G'lv‘lVlwl‘ ATTTAAAACTGTGGACTTAGCATJMXAAAGGGAGACTATATTTATTTTTTACA AAAGGGA'I‘AAAAATGGAACCC'l"l"l‘CCTCACCCACCAGA’I‘Tl‘AG'l‘CAGAAAAA AACATTCTATTCTGAAACSGTCACAGTGGTTTTGACATGACACATCAGAACAA CGCACACTGTCCATGA’I’GGCTrl‘A’l‘GAAC'i‘CCAAGTCACTCCr—‘i’l‘CA’l’GG’l’AAJ-‘k TGGGTAGATCCCTCCTTCTAGTGTGCCACACCATTGCTTCCCACAGTAGAATC ’l‘TAT’l‘TAAG’l‘GC’I‘AAGrl‘GTTG'i‘CTCTGC’l‘GG'i‘TTAC’l‘CTGT"l‘GTTTl‘AGAGAA’l‘ GTAAGTTGTATAGTGAATAAGTTATTGAAGCATOTGTAAACACTGTTATACA TCTTTTCTCCTAGATGGGGAATTTGGAATJMXAATACCTTTAAAATTCAAJMXA AAAAAAAAAAAAA Karma nervegicus C9erf72 amine acid (NFjlitiitiW’YlB; SEQ 1D N0:4) CPFPSFAVAK’I‘ElALSGFSPLLAATFAY‘WDNlLGFR‘VRHl‘WAPK’l‘flQVLLS DGElTFLANE-lTLNGElLWAESGAlDVKFFVLSEKGVHVSLIFDGNWNGDRSTYG LSllLPQTELSEYLPLHRVCVDRLTHHRKGRIWMHKERQENVQKlVLEGTERIVlED QGQSHPMLTGEVIPVMELLASMRSHSVPEDLDlADWLNDDDIGDSCH-1136FLLN AlSSHLQTCGCSVVVGSSAEKVNEGVR’I‘LCLFL'i‘l’AERlQCSRLCEAESSFl‘LYESGL FVQGLLKDATGSFVLPFRQVMYAFYPTTi-i{DVDVNFVKQMFPCHEHHNQREY AFW’RATSEEDMAQDTHYTDESFTPDLNIFQDVLl-iRDTLVKAFLDQVFH LKPGLS1RSTFLAQFLLILHRKAL’l‘LlKYlEI)DTQKGKKPFKSLRNLKIDLl)lfl‘Ali GDLNllMALAEKlKPGLHSFlFGRPFYTSVQERDVLMTF {5901?}??? Targeting Waters and Predactien af'Nmt—Human s Having a Disruptinn in a C90RF72 Lawns {wait} Provided herein are targeting vectors or targeting ucts fer the productien of non—human animals having a disruption in a C9QRF72 locus as described herein.
EiltilEll} DNA sequences can be used in prepare targeting veetcrs fer kneekeut animals (fag, an C90RF72 K0). Typically, a. pclynnclectide niclecnle (egg, an insert c acid) encoding a er gene and/er a selectable marker is inserted into a vectcr, preferably a DNA vecter, in order tc replicate the pelynuclectide ule in a suitable host cell, fitililfii} A pelynuclectide inelecule (0r insert nucleic acid) comprises a segment of DNA that one desires to integrate into a target locus. in some embodiments, an insert nucleic acid cernprises cne er rncre pelynuclectides Of interest. in seine enibcdirnents, WO 96185 an insert nucleic acid comprises one or more expression cassettes. in some certain embodiments, an expression cassette comprises a cleotide of interest, a polynucleotide encoding a selection marker and/or a reporter gene along with, in some certain ments, various regulatory components that influence expression.
Virtually any poly/nucleotide of interest may be contained within an insert nucleic acid and thereby integrated at a target genomic locus. Methods disclosed herein, provide for at least l, 2, 3, 4, 5, 6 or more polynucleotides of interest to be integrated into a targeted C9GRF72 genomic locus. {@9132} In some embodiments, a polynucleotide of interest contained in an insert nucleic acid encodes a reporter. In some embodiments, a cleotide of interest encodes a selectable inarleert {@9133} in some embodiments, a poly/nucleotide of interest is flanked by or ses site-specific recombination sites (e.g., ioxP, Frt, etc). in some certain embodiments, pecific recombination sites flank a DNA t that encodes a reporter and/or a DNA segment that s a selectable marker. Exemplary polynucleotides of interest, including selection s and reporter genes that can be included within insert c acids are described , {$6134} Various methods employed in preparation of plasmids, DNA constructs and/or targeting s and transformation of host organisms are known in the art. For other suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning: A Laboratory Manual, 2nd Ed, ed. by Sambrook, L et alt, Cold Spring Harbor Laboratory Press: l989. {@135} As described above, exemplary non-human (cg, rodent) C90RF72 nucleic acid and amino acid sequences for use in constructing targeting vectors for ut animals are provided in Table lo Other non—human CQORF72 sequences can also be found in the GenBanl: database. CWHlF172 targeting vectors, in some embodiments, comprise DNA sequences encoding a reporter gene and/or a selectable marker, flanked by sequences that are identical or substantially homologous to llanking sequences of a target region (also referred to as "homology arms”) for insertion into the genome of a transgenic non~human animal. To give but one e, a deletion start point may be set upstream (5') of a first exon, a first coding exon, or the first or second codon, to allow an insert nucleic acid to be operably linked to an endogenous regulatory sequence (eg, a promoter). Figure 1A illustrates a ing strategy for making a targeted deletion of the entire coding sequence of a mur'ine C90rgf72 gene and replacement with a te that contains a sequence from a [6202’ gene that encodes B—galactosidase and a drug selection cassette (Neo) that encodes neonrycin phosphotransferase for the selection of G418" resistant embryonic stem (ES) cell colonies. The targeting vector also es a sequence encoding a recombinase (eg, Cre) regulated by an ES—cell specific miRNAs or a germvcell ic promoter (e.g., protamine l promoter; Prot—CreSV40). The drug selection cassette and Cre recombinase—encoding ces are flanked by Zoxl’ (LP) recombinase recognition sites that enable Cre~mediated excision of the drug selection cassette in a pment~dependent mariner, e.g., progeny derived from rodents whose germ cells containing the ted ?? gene described above will shed the selectable marlrer (Neo) during development {see US. Patent No.'s 8,697,85 l, 8,5l3,392, 8,354,389, 8,946,505, and 8,946,504, all of which are herein incorporated by reference).
This allows for, among other things, automatic excision of the selection cassette from either ditlerentiated cells or germ cells. Thus, prior to phenotypic analysis the drug selection cassette is removed leaving only the ZacZ er gene operably linked to the murine CQQWZ promoter. fiblllfidl As bed herein, disruption of a C90rf72 locus can comprise a replacement of or an insertion/addition to the C79orgf‘72 locus or a portion thereof with an insert nucleic acid. in some embodiments, an insert c acid comprises a reporter gene. in some certain embodiments, a reporter gene is oned in le linkage with an endogenous C9orjf72 promoter. Such a modification allows for the expression of a reporter gene driven by an endogenous C905f‘72 promoter. Alternatively, a reporter gene is not placed in operable linkage with an endogenous Clint/fir? promoter. fiblllfi’l’l A. variety of reporter genes (or detectable moieties) can he used in targeting vectors described herein. Exemplary reporter genes include, for example, {in osidase (encoded £7:ch gene), Green Fluorescent Protein (GFP), enhanced Green Fluorescent Protein (eGFF), MmGFl’, blue fluorescent n (EFF), enhanced blue scent protein (eBFP), anlurn, mCherry, thomato, in Strawberry, l—Red, DsRed, mOrange, niKO, ine, Venus, Yl’et, yellow fluorescent protein (’YFP), enhanced yellow fluorescent protein (eYFl’), Emerald, Cyl’et, cyan fluorescent protein (CFP), Cerulean, T—Sapphire, lucil’erase, alkaline phosphatase, or a combination f. The methods described herein trate the construction of targeting vectors that employ the use of a lad? reporter gene that encodes ligalactosidase, however, persons of skill upon g this disclosure will understand that non—human animals bed herein can be generated in the absence of a reporter gene or with any reporter gene known in the art. {$6138} Where appropriate, the coding region of the genetic material or polynucleotide sequence(s) encoding a reporter polypeptide, in whole or in part, may be modified to include codons that are zed for expression in the man animal (eg, see US, Patent No.'s 5,670,356 and 5,874,304), Codon optimized sequences are synthetic sequences, and preterably encode the identical polypeptide (or a ically active fragment of a full length polypeptide which has substantially the same activity as the full lengh polypeptide) d by the non~codon optimized parent poly/nucleotide. in some embodiments, the coding region of the genetic material encoding a reporter ptide (e.g. [6202}, in whole or in part, may include an altered sequence to optimize codon usage for a ular cell type (eg, a rodent cell). For example, the codons of the reporter gene to be inserted into the genome of a man animal (eg, a rodent) may be zed for expression in a cell of the nonnhuman animal. Such a sequence may he described as a codonuoptimized sequence. {@9139} Compositions and methods for making non—human animals that comprises a. disruption in a C9GRF72 locus as described herein are provided, including compositions and methods for making man animals that express a reporter gene from a C90RF172 promoter and a C90RF72 regulatory sequence. In some ments, compositions and methods for making nonrhuman animals that express a reporter gene from an endogenous promoter and an endogenous regulatory sequence are also provided, Methods include ing a ing vector, as described herein, encoding a reporter gene (eg, lacZ) into the genome of a non~hurnan animal so that an entire coding sequence of a (7901231972 locus is deleted, in Whole or in part, ln some embodiments, methods include inserting targeting vector into the genome of a nonuhuman animal so that the entire coding sequence for all C90RF172 isotorrns at a C9013F72 locus is deleted. ltltlldtl} Insertion of a reporter gene operably linked to a C9QRF72 promoter (e.g., an endogenous C90RF72 promoter) employs a relatively minimal modification of the genome and results in expression of reporter polypeptide in a C90RE‘72—specific manner in the non—human animal. in some embodiments, a nonnhuman animal bed herein comprises a C90RF72 locus that comprises a targeting vector as described herein. 399141} Targeting vectors described herein may be introduced into ES cells and screened for ES clones harboring a disruption in a CQorj’W locus as described in Erendewey, ll, et al,, 20w, Methods Enzymol, 476295—307. A variety of host embryos can be employed in the methods and compositions disclosed herein. For example, the pluripotent and/or totipotent cells having the ed genetic cation can be introduced into a pre~rnorula stage embryo (eg, an S—cell stage ) from a corresponding organism. See, egg US 7,5 76,259, US 442, US 7,294,754, and US zoos/007mm Al all of which are incorporated by reference herein in their entireties, in other cases, the donor ES cells may be implanted into a host embryo at the 2—cell stage, 4—cell stage, S-cell stage, lo-celi stage, 32~cell stage, or nit-cell stage. The host embryo can also be a blastocyst or can be a prenhlastocyst embryo, a pre—morula stage embryo, a mornla stage embryo, an uncompacted morula stage , or a compacted morula stage embryo. [hilldzl in, some embodiments, the VELOCIMOUSEQ?) method (Poucymirou, Will et al.,, 2007, Nat. Biotechnol. 25:9ln99) may he d to inject positive ES cells into an 8- cell embryo to generate fully ES cell—derived F0 generation heterozygous mice ready for ZacZ expression profiling or breeding to homozygosity. Exemplary methods for generating non—human animals having a disruption in a C90rf72 locus are provided in Example l. {@9143} s for ting transgenic non—human animals, including uts and knockins, are well known in the art (see, eg, Gene 'l‘argeting: A Practical Approach, Joyner, edo, le‘ord sity Press, inc, (2000)). For example, generation of transgenic rodents may optionally involve disruption of the genetic loci of an endogenous rodent gene and introduction ofa er gene into the rodent genome, in some embodiments, at the same location as the endogenous rodent gene. [dill/Ml A tic illustration {not to scale) of the genomic organization of a mouse C9005? is provided in Figure lA. An exemplary targeting strategy for on of an entire coding sequence of murine C90rjf72 locus using a reporter gene is also provided in Figure lA, As illustrated, genomic DNA containing the coding portion of exon 2 through the coding portion of exon ll ot‘a marine C9riigf72 locus is deleted and replaced with a reporter gene and a drug selection cassette flanked by site—specific recombinase recognition sites, The targeting vector used in this strategy includes a recombinasen encoding sequence that is ly linked to a promoter that is developmentally regulated such that the reeoinbinase is sed in undifferentiated cells. Exemplary ers than can be included in targeting vectors described herein are provided in 'l'able 2. Additional suitable promoters that can be used in targeting vectors bed herein include those described in US. Patent N033 8,697,851, 8,518,392 and 8,354,389; all ot‘whieh are herein incorporated by reterenee). Upon homologous recombination, the entire coding sequence (e.g., the coding portion of exon 2 through the coding portion of exon l l) of an nous rnurine CQQIflZ locus is replaced by the sequence contained in the targeting vector. The drug selection cassette is removed in a development— riependent manner, i.e., progeny d front niice whose germ line cells containing a disruption in a C90zf72 locus described above will shed the selectable marker front difterentiated cells during development (see US Patent Nofs 8,697,851, 8,5i8,392 and 8,354,389, all of which are herein incorporated by reference). l’rot er {SEQ ll“) Nflzfi} CCAG’I‘AGCAGCACCCACGTCCACC’l‘TC’l‘G’l‘C’l‘AGTAATG'l‘CCAACACCTCCCT CACS’TCCAAACACTGCTCTGCATCCATGTGGCTCCCATTTATACCTGAAGCACT l"(EGGGCC"i‘CAA’l‘G’l‘T’i"l‘ACTAGAGCCCACCCCCC’l‘GCAAC’l‘C’l‘GAGACC CTCTGGATTTGTCTGTCAGTGCCTCACTGGGGCGTTGGATAATTTCTTAAAAG GTCAAGTTCCCTCAGCAGCA’l‘TC’l‘C’l“GAGCAGTC’l‘GAAGA’l‘GTG’i‘GC’l"l"i"l‘CA CAGTTCAAATCCATGTGGCTGTTTCACCCACCTGCCTGGCCTTGGGTTATCTA CCTAGCC’l‘AGAAGCAGG’l‘G’l‘G’l‘GGCAC’l‘l‘AACACC’l‘AAGC’i‘GAG’l‘G ACTAACTGAACACTCAAGTOGATGCCATCTTTGTCACTTCTTGACTGTGACAC AAGCAAC’l‘CCTGA’l‘GCCAAAGCCCTGCCCAC‘.CCC’l‘C’l‘CA’l‘GCCCA’l‘AT’l‘TGG ACATGGTACAGGTCCTCACTGGCCATGGTCTGTGAGGTCCTGGTCCTCTTTGA CTTCiI-‘i'l‘itn/fi'lTCC’l‘AGGGGC(SAC’l‘AG’l‘A’l‘C’l‘A’i‘iMXGAGGAAGAGGG’l‘GCTGGC TCCCAGGCCACAGCCCACAAAATTCCACCTGCTCACAGGTTGGCTGGCTCGA CCCAGG’lGG’l‘GTCCCC’l‘GC’l‘C’l‘GAGCCAGC’l’CCCGGCCAAGCCAGCACC Blimpl promoter lkb (SEQ ll) N026) 'l'GCCA’l'CATC.ACAGGA'l‘G'l‘CC’l‘l‘CC’l‘l‘C’l‘CCAGAAGACAGAC'l'GGGGCTGAA GGAAAAGCCGGCCAGGCTCAGAACGAGCCCCACTAATTACTGCCTCCAACA GCT'l‘TC-CACPl‘CAC'l‘GCCCCCAGCCCAACATCCCC'I‘T'l'T'l‘AAC'l‘GGGAAGCA'l"l' CCTACTCTCCATTG’TACGCACACGCTC(SGAAGCCTGGCTGTGG(S'TTTCSGGCA 'l'GAGAGGCAGGGACAACAAAACCAG’FATATA'l‘GA’l"l'ATAAC'I'"i"l"l"l‘CCTGTT TCCCTATTTCCAAATGGTCGAAAGGAGGAAGTTAGGTCTACCTAAGCTGAAT STAT'l‘CAGl‘TAGCAGGAGAAATGAAA’l‘CCTA’l‘ACG’l‘l‘TAATACTAGAGGAGA ACCGCCTTAGAATATTTATTTCATTGGCAATGACTCCAGGACTACACAGCGA 2016/034304 AATTGTA’I‘TGCATGTGC’E‘GCCAAAA’I’ACTTI’AGC'I’CT’I‘TCC’I‘TCGAAGTACGT CGGATCCTGTAATTGAGACACCGAGTTTAGGTGACTAGGGTTTTCTTTTGAG GAGGAG’E‘CCCCCACCCCGCCCCGCTC’I‘GCCGCGACAGGAAGCTAGCGATCCG GAGGACTTAGAATACAATCGTAGTGTGGGTAAACATGGAGGGCAAGCGCCT GCAJMXGGGAAGTAAGAAGA’I’TCCCAGTCCT’E‘GITGAAA’E‘CCA’I‘TTGCAAACA GAGGAAGCTGCCGCGGGTCGCAGTCGGTGGGGGGAAGCCCTGAACCCCACG CTGCACGGCTGGGCTGGCCAGG’I’GCGGCCACGCCCCCATCGCGGCGGCTGGT AGGAGTGAA'E‘CAGACCGTCAG'I'ATI‘GG’I‘AAAGAAG'E‘CTGCGGCAGGGCAGG GAGGGGGAAGAGTAGTCAGTCGCTCGCTCACTCGCTCGCTCGCACAGACACT GCTGCAGTGACACTCGGCCCTCCAGTGTCGCGGAGACGCAAGAGCAGCGCG CACS’CACCTG’TCCGCCCGGAGCGAGCCCG’GCCCGCGGCCGTAGAAAACS’GAGG GACCGCCGAGGI‘GCGCGTCAGTAC'I‘GCTCAGCCCGGCAGGGACGCGGGAGG ATGTCS’GACTGGGTGGAC Biimpl pmmater Zkh (SEQ ID N027) GTGG’I‘GCTGACTCAGCATCGGI‘TAATI'AAACCCTC’E‘GCAGGAGGC’E‘GGATTTC TTTTGTTTAATTATCACTTGGACCTTTCTGAGAACTCTTAAGAATTGTTCATTC TTT’E"1TGT’I‘TTG’I‘TTTGG’I’T’I‘GGTTT’I‘TTTGGG’I"1"1TT’1"1"1‘TT’I‘TTT’E‘TTTT TTTGGTTTTTGGAGACAGGGTTTCTCTGTATATAGCCCTGGCACAAGAGCAA {3C’I‘AACAGCCTGTT’E‘C’ITC’I‘TGGTGCTAGCGCCCCCTCTGGCAGAAAATGAA ATAACAGGTGGACCTACAACCCCCCCCCCCCCCCCCAGTGTATTCTACTCTTG TCCCCGGTA’I‘AAAT’I’TGATTG’I’TCCGAACTACATAAA’I"TGTAGAAGGATTTTI‘ TAGATGCACATATCATTTTCTGTC:ATACCTTCC.ACACACCCCTCCCCCCCAAA ‘MXAATT’ITTCTGGGAAAG’ITI‘CT”E‘GAAAGGAAJMXCAGAAGAACAAGCCTG’E‘C TTTATGATTGAGTTGGGCTTTTGTTTTGCTGTGTTTCATTTCTTCCTGTAAACA AATAC’E‘CAAATGTCCACT?CATTG’I‘A’I’GAC’I’AAGTTGGTATCAT’I‘AGGTI’GGG TCTGGGTGTGTGAATGTGGGTGTGGATCTGGATGTGGGTGGGTGTGTATGCC GTTTAGzMU‘AC’I‘AGAI’XAAGATACCACATCGTAAACT’E‘TTGGGAGAG A'I‘GA'i"I"i"TTAAAAATGGGGG’I‘GGGGGTGAGGGGAACCTGCGATGAGGCAAG CAAGATAAGGG(SAAGACTTGAGTTTCTGTGATCTAAAAA(S'TCGCTGTGATGG GATGCTGGCTA’E‘AAATGGGCCCTTAGCAGCA'I‘TGTI‘TC“E‘G'I‘GAATTGGAGGA TCCCTGCTGJMXGGCAAAAGACCATTGAx—‘XGGAAGTACCGCATCTGGTTTGTTT TG’I‘AA’I‘GAGAAGCAGGAATGCAAGGTCCACGCTCTTAATI'AATAAACAAACA GGACATTGTAT(3CCATCATCACAiKiATGTCCTTCCTTCTCCACS’AAGACAGAC TGGGGC’I'GAAGGAAAAGCCGGCCAGGC’I'CAGAACGAGCCCCACTAA’ITACT (3CCTCCAACAGCTTTCCACTCACTGCCCCCAGCCCAACATCCCCTTTTTAACT GGGAAGCA'E‘TCC’E‘ACTCTCCATTGTACGCACACGCTCGGAAGCC'I‘GGCTGTG GGTTTGG(SCATGAGAGGCAGGGACAACAAAACCAGTATATATGATTATAAC T’I’TTTCCTG’I‘TTCCC'i‘A’l"I"i‘CCAAATGGTCGAAAGGAGGAAGTTAGGTCTACC TAAGCTGAATGTATTCAGTTAGCAGGAGAAATGAAATCCTATACGTTTAATA CTAGAGGAGAACCGCCT’I‘AGAA’EYU"1"1‘A’l"I‘TCA’E"I‘GGCAATGACTCCAGGAC TACACAGCGAAATTGTATTGCATGTGCTGCCAAAATACTTTAGCTCTTTCCTT CGAAGTACGTCGGATCC’I’GTAAT’E‘GAGACACCGAGTTTAGGTGAC”E‘AGGGTT TTCTTTTGAGGAGGAGTCCCCCACCCCGCCCCGCTCTGCCGCGACAGGAAGC ’I’AGCGA’E‘CCGGAGGACTTAGAATACAATCG’i‘AG’E‘GTGGG’E‘AAACA’I‘GGAGG GCCTGCAAAGGGAAGTAAGAAGATTCCCAGTCCTTGTTGAAATCCA T’I’TGCAAACAGAGGAAGCTGCCGCGGGTCGCAGTCGGTGGGGGGAAGCCCT GAACCCCACGCTGCACGGCTGGGCTGGCCAGGTGCGGCCACGCCCCCATCGC Gii’CGGCTGGTAGGAGTGAATCAGACCG’TCACS’TATTGGTAAAGAAGTCTGCG GCAGGGCAGGGAGGGGGrM‘aGAG’l‘AG’l‘CAGTCGCTCGC'l‘CACTCGC'l’CGC'l‘C GCACAGACACTGCTGCAGTGACACTCGGCCCTCCAGTGTCGCGGAGACGCAA CGCGCAGCACCTGTCCGCCCGGAGCGAGCCCGGCCCGCGGCCGTA GAAAAGGAGGGACCGCCGAGGTGCGCGTCAGTACTGCTCAGCCCGGCAGGG ACGCGGGAGGATGTGGAC’l‘GGGTGGAC } A transgenic founder non-human animal can be identified based upon the presence ot‘a reporter gene (or absence ofCQORWZ) in its genome and/or expression of a reporter in tissues or cells of the non—human animal (or lack of expression of C90RF72). A, enic founder non—human animal can then be used to breed additional non~human animals carrying the reporter gene thereby creating a series of non~human animals each carrying one or more copies of a CQORF72 locus as described herein. 399146} Transgenic nonnhuman s may also be produced to contain selected systems that allow for regulated or directed expression of the transgene. Exemplary systems include the tire/20x? recombinase system of bacteriophage Pl (see, e.g., Lahsc, M. et al., l992, ?roc Natl. Acad. Sci. USA 89:6232—6236) and the FlJP/Frt inase system of S. cereyislae (Q’Gorrnan, S. et al, l99l , Science 25l : l 35l—l355). Such animals can be ed through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected polypeptide (eg, a reporter gene) and the other containing a transgene encoding a recombinase (e.g., a Cre recombinase}. ltlllld'l’} Although embodiments employing a disruption in a C90RF172 locus in a mouse (i.e., a mouse with a on of an entire 72—coding sequence) are extensively discussed herein, other non—human s that comprise a disruption in a C90RF172 locus are also provided. in some embodiments, such non—human animals se a disruption in a C901RF72 locus characterized by insertion of a reporter operably linked to an nous C90RF72 promoter, Such nonmhuman animals include any of those which can be cally modified to delete an entire coding sequence of a C90RF72 locus as sed herein, including, cg, mammals, e.g., mouse, rat, rabbit, pig, bovine (cg, cow, bull, buffalo), deer, sheep, goat, chicken, cat, dog, ferret, e (e.g., marmoset, rhesus monkey), etc. For example, for those non— human animals for which suitable genetically modifiable ES cells are not readily available, other methods are employed to make a nonvhurnan animal comprising the genetic modification. Such methods include, e.g,, modifying a non—ES cell genome (cg, WO 96185 a ast or an induced pluripotent cell) and employing somatic cell nuclear transfer (SCNT) to transfer the genetically modified genome to a suitable cell, e. 9;, an ated oocyte, and gestating the modified cell leg, the modified oocyte) in a non—human animal under suitable conditions to form an embryo. {@9148} Briefly, methods for nuclear transfer include steps of: (l) enucleating an oocyte; ('2) isolating a donor cell or s to be combined with the enucleated oocyte; (3) inserting the cell or nucleus into the enucleated oocyte to form a reconstituted cell; (4) implanting the reconstituted cell into the womb of an animal to form an embryo; and (5) allowing the embryo to develop. in such methods oocytes are generally retrieved from deceased s, gh they may be isolated also from either oviducts and/or ovaries of live animals. Oocytes may be matured in a variety of medium known to persons of skill in the art prior to enucleation. Enucleation of the oocyte can be performed in a variety of ways known to persons of skill in the art. insertion of a donor cell or nucleus into an ated oocyte to form a reconstituted cell is typically ed by microinjection ofa donor cell under the zona pellucida prior to fusion. Fusion may be induced by application ot‘a DC electrical pulse across the contact/fitsion plane (electrofusion), by exposure of the cells to -promoting chemicals, such as polyethylene , or by way of an inactivated Virus, such as the Sendai virus. A reconstituted cell is typically activated by ical and/or ectrical means before, during, and/or alter fusion of the nuclear donor and recipient oocyte. Activation methods include electric pulses, chemically induced shoclr, penetration by sperm, increasing levels of divalent cations in the oocyte, and reducing orylation of cellular proteins (as by way ot‘kinase inhibitors) in the oocyte. The activated reconstituted cells, or embryos, are typically cultured in medium known to persons of skill in the art and then transferred to the womb of an animal. See, e.g., LES. Patent Application Publication No. 200843092249 Al W0 l999/(lll5266 A2, US. Patent Application Publication No. 2004" (ll 773% Al, WO ZQGS/Ol 7234 Al, and U.S. ?atent No. 7,6l2,’250, each of which is herein incorporated by nce. £99149} Methods for modifying a nonnhuinan animal genome leg, a pig, cow, rodent, n, etc.) include, e.g., employing a zinc linger nuclease (ZFN) or a transcription activator-like effector nuclease ('l‘ALEN) to modify a genome to include a disruption in a (39013572 locus as described herein. fiddlfilll in some ments, a non~hnman animal described herein is a mammal.
In some embodiments, a man animal described herein is a small mammal, eg., or“ the snperfamily litipodoidea or Moroidea. in some embodiments, a genetically modified animal described herein is a rodent. in some embodiments, a rodent described herein is selected from a mouse, a rat, and a hamster. In some ments, a rodent described herein is selected born the snpertamily Moroidea. in some embodiments, a c lly ed animal described herein is from a family selected from Calomyscidae {e.g,, moase~lilce hamsters), idae (egg hamster, New World rats and mice, voles), Mnridae (true mice and rats, gerbils, spiny mice, crested rats), Nesomyidae (climbing mice, rock mice, with—tailed rats, Malagasy rats and mice), antbomyidae leg, spiny dormice), and Spalacidae leg, mole rates, bamboo rats, and zohors). in some n embodiments, a genetically moditied rodent described herein is selected trom a true mouse or rat (family Maridae), a gerbil, a spiny mouse, and a crested rat. in some certain embodiments, a genetically d mouse described herein is from a member of the family Mnridae. in some embodiment, a non—lniman animal described herein is a rodent. In some certain embodiments, a rodent described herein is selected from a mouse and a rat. in some embodiments, a non—human animal described herein is a mouse. [0015ll in some embodiments, a nonuhuman animal described herein is a rodent that is a mouse ofa C5TEL strain selected from CS7BL/A, C5 ”ibis/An, C57BL/Grl7a, C57BL/Kaliwbl, C57BL/d, C5 7BL/6J, C578L/68y5, cysts/{5N}, C57Bli/l 0, C573L/l0Se-Sn, CS7BL/lGCr, and C57BL/Ola. in some certain embodiments, a mouse described herein is a l29 strain selected from the group consisting ot‘a strain that is 129Pl, l29P2, l29P3, lZQXl, l29$l (ea, SV, lZQSl/Svlm), 12952, 12984, 129S5, lZQSQ/SVEVH, lZ9/Svjae, 12986 (129/SVEV’l‘ac), l29S7, lZQSS, l29'i‘l, l29’l‘2 (see, e.g., Festing et al., l999, Mammalian Genome l0:336; Anerbaoh, W. et al, 2000, Biotechniques 29(5): l024—1028, l030, l032). In some certain embodiments, a cally modified mouse described herein is a min of an aforementioned l29 strain and an aforementioned C57BL/6 strain. in some certain embodiments, a mouse described herein is a mix of aforementioned l29 strains, or a mix ementioned Eli/{i strains. in some certain embodiments, a L29 strain of the mix as described herein is a l2986 {lZQ/Svliy’l‘ac) strain. in some embodiments, a mouse described herein is a BALE strain, e.g, BALE/c strain, in some embodiments, a mouse described herein is a mix ofa BALE strain and another aforementioned strain. {@9152} in some embodiments, a man animal described herein, is a rat, in some certain embodiments, a rat bed herein is selected from a Wistar rat, an LEA strain, a Sprague Dawley strain, a Fischer strain, F344, F6, and Dark Agouti. in some certain embodiments, a rat strain as bed herein is a mix of two or more strains selected item the group consisting of Wistar, LEA, e Dawley, Fischer, F344, F6, and Dark Agcuti. {@9153} A rat plurinctent and/or tctipotent cell can be from any rat strain, including, for example, an AC1 rat strain, a Dark Agouti (DA) rat strain, a Wistar rat , a LEA rat , a Sprague Dawley (SD) rat strain, or a Fischer rat strain such as Fisher F344 or Fisher F6. Rat nlurinotent and/or totipotent cells can also be obtained from a strain derived from a mix cftwo or more strains recited above. For e, the rat pluripotent and/or totipotent cell can be item a DA strain or an ACl strain. The AC1 rat strain is characterized as having black agcuti, with White belly and feet and an RN” haplotype. Such strains are available from a variety of sources including, an Lahoratcries, An example of a rat ES cell, line item an ACl rat is an ACLGl rat ES cell.
The Dari; Agouti (DA) rat strain is characterized as having an agouti coat and an RN” ype. Such rats are available from a variety of sources including Charles River and Harlan Lahoratcries, Examples ofa rat ES cell line from a DA rat are the DAEB rat ES cell, line and the DAQC rat ES cell line. in some cases, the rat pluripotent and/or totipotent cells are from an inbred rat strain, See, e.g., US, 2014/0235933 Al, filed on February 20, 20l 4, and herein ineorpcrated by nce in its entirety.
Elllllfidl Non-human animals are provided that se a disrupticn in a CQORF72 locus, in some embodiments, a disruption in a C90RF72 locus results in a loss-cf— cn. in particular, loss—ofui’unction mutations include mutations that result in a decrease cr lack of expression ofCEPQRFH and/or a decrease or lack of activity/function of C90RF72. in some embodiments, loss—of—fimction mutations result in one or more phenotypes as described herein, Expression of (3903572 may be measured directly, e.g., by assaying the level of CQORF72 in a cell or tissue of a non—human animal as described herein, {$6155} Typically, expression level and/or activity of CTQGRE‘T’Z is decreased if the expression and/or ty level of C9ORF72 is statistically lower (950.895) than the level ol‘CQORP72 in an appropriate control cell or non—human animal that does not comprises the same disruption {e.g., deletion). In some embodiments, concentration and/or activity of CQORFT’IZ is decreased by at least i “/0, 5%, l0%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more relative to a control cell or non~hurnan animal which lacks the same disruption {e.g., deletion). {39156} in other embodiments, cells or organisms having a disniption in a C90RF72 locus that reduces the expression level and/or activity of C90RF72 are selected using methods that e, but not limited to, Southern blot analysis, DNA sequencing, PCR analysis, or phenotypic is. Such cells or man animals are then employed in the various methods and compositions described . {$6157} in some embodiments, an nous (39012572 locus is not deleted (i.e., intact). in some embodiments, an endogenous C90RF72 locus is altered, disrupted, deleted or replaced with a heterologous sequence (eg, a reporter gene encoding sequence), in some embodiments, all or ntially all of an endogenous CQORF72 locus is replaced with an insert nucleic acid; in some certain embodiments, replacement includes replacement of an entire coding sequence of an endogenous (3901117772 locus with a lad? er gene so that the lack? er gene is in operable linkage with a (39013572 promoter (egg an endogenous (79031472 promoter), in some embodiments, a portion ot‘a reporter gene (e.g., a function nt thereof) is inserted into an nous non—human C9GRF72 locus. in some embodiments, the reporter gene is a Incl gene. in some ments, a reporter gene is ed into one of the two copies of the endogenous (39013572 locus, giving rise to a nonnhuman animal that is heterozygous with respect to the reporter gene. in some embodiments, a non—human animal is provided that is homozygous for a reporter gene.
Methods Employing Nonmhnman Anirnals Having Disruption in o C983F713 Locus } Non—human animals as described herein provide improved animal models for neurodegenerative diseases, disorders and conditions. In particular, non—human animals as described herein provide improved animal models that translate to human diseases such as, for example, ALS and/or F'l'l'), characterized by upper motor neuron symptoms and/or non~rnotor neuron loss. {@159} For example, a disruption in a C90RF172 locus as described herein may result in s symptoms (or phenotypes) in the non—human animals ed herein, In some embodiments, deletion of a C9012F72 locus results in man animals that are grossly normal at birth, but that develop ALSulike symptoms upon aging, eg, alter about 8 weeks, 9 weeks, lll weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, 23 weeks, 29 weeks, 30 weeks, 3i weeks, 32 weeks, 33 weeks, 34 weeks, 35 weeks, 36 weeks, 37 weeks, 38 weeks, 39 weeks, 40 weeks, 41 weeks, 42 weeks, 43 weeks, 44 weeks, 45 weeks, 46 weeks, 47 weeks, 48 weeks, 49 weeks, 50 weeks, 51 weeks, 52 weeks, 53 weeks, 54 weeks, 55 weeks, 56 weeks, 57 weeks, 58 weeks, 59 weeks, 60 weeks, etc. 1n some embodiments, on of a Z‘ locus results in abnormal functions of one or more cell types, e.g., a neuron and/or a portion thereof, A neuron includes a sensory neuron or a motor neuron. Other phenotypes associated with ALS and/or FTD may be present in inan animals described herein. For example, an ALB—like phenotype may involve impairment of one or more neurons, e,g,, motor neurons and/or sensory neurons, Further, an Al..S—lil<e phenotype involving upper motor neurons may result in spastieity , spastie paralysis, ty), increased and/or abnormal reflexes (eg, Bahinski’s sign), tremors and a combination thereof. An ALS-like phenotype inyolying impairment of lower motor neurons may result in muscle weakness and wasting, t‘asoieulations, and a combination thereof, and/or ment of the bulbar resulting in an inability to swallow and tongue fasciculations. Ah ALS—like symptom may also comprise one or more of the following phenotypes: a) kyphosis; h) abnormal hind limb elasping, dragging or toe curling; c) defieienoy in motor coordination and, motor learning ability, deficiency in rotarod, k and/or open field testls); d) motor neuron loss in the spinal cord; e) astrocytosis in the spinal cord; t) weight loss compared with a control rodent; g) accumulation of hiquitinated proteins and/or (h) increased ogical scoring using the ALSn’l‘Dl neurological scoring system (Table 3).
TABLE 3 ALS—E‘Dl neurological scoring system Score of l): Full extension of hind legs away from lateral rnidline when mouse is suspended by its tail, and mouse can hold this for two seconds, suspended two to three times. 2016/034304 Score of l: Collapse or partial collapse of leg extension towards lateral midline (weakness) or ing ot‘ hind legs during tail suspension.
Score of 2: "l‘oes curl under at least twice during walking of l2 inches, or any part of foot is dragging along cage bottom/tablet Score of 3: Rigid paralysis or minimal joint movement, foot not being used for generating forward motion.
Score of 4: Mouse cannot right itself within 30 seconds after being placed on either side, {auras} Thus, in at least some embodiments, non-human animals described herein provide improved animal models for neurodegenerative diseases, ers or conditions (cg, ALS and/or ETD) and can be used for the development and/or identification of therapeutic agents for the treatment, tion and/or inhibiting one or more phenotypes (or symptoms) of neurodegenerative diseases, disorders or conditions. ln some ments, one or more symptoms (or phenotypes) in non—human animals described herein appear in Table 3. [llllléll Non-human animals as described herein also provide an in viva system for identifying a therapeutic agent for treating, preventing and/or inhibiting one or more symptoms ofneurodegenerative diseases, disorders or conditions (eg, ALS and/or ETD). in some embodiments, an inhibitory effect of a therapeutic agent is determined in viva, by stering said therapeutic agent to a nonvhuman animal that has a (790RF72 disruption as described , and develops egenerative symptoms alter 38 weeks of age. {@162} Non-human animals as described herein also provide ed animal models for inllannnatory or autoimmune diseases, disorders and conditions. in ular, non—human animals as bed herein provide improved animal models that translate to human inflammatory disease characterized by infiltration of immune cells in various organs (eg. kidney, liver, spleen, etc.). in addition, nonmhuman animals as described herein provide improved animal models that translate to human autoimmune disease characterized by the increased presence oi‘autoantibodies (e.g., lgG and lglvl) in the serum, } For example, a disruption in a 6'9031‘72 locus as described herein may result in various conditions (or phenotypes) in the nonuhuman animals provided herein. in some ments, on of a (390121472 locus results in man animals that are grossly normal at birth, but that develop inflammatory and/or mune conditions upon aging, eg., after ahout 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, 23 weeks, 29 weeks, 30 weeks, 3l weeks, 32 weeks, 33 weeks, 34 weeks, 35 weeks, 36 weeks, 37 weeks, 38 weeks, 39 weeks, 40 weeks, 41 weeks, 42 weeks, 43 weeks, 44 weeks, 45 weeks, 46 weeks, 47 weeks, 48 weeks, 49 weeks, 50 weeks, 51 weeks, 52 weeks, 53 weeks, 54 weeks, 55 weeks, 56 weeks, 57 weeks, 58 weeks, 59 weeks, 60 weeks, etc. in some embodiments, deletion of a C90RF172 locus results in an infiltration of one or more immune cell types, eg., plasma cells, monocytes, granuloeytes and/or macrophages, Other phenotypes associated with an atory and/or autoimmune condition may he present in non—human s described herein, For example, an inflammatory or autoimmune condition may involve enlargement of one or more of the spleen, lymph nodes, kidney, and/or liver. Further, an inflammatory or autoimmune condition involving the blood may result in an increase presence of autoantihodies. An inflammatory or autoimmune eondition involving the liver may result in hepatitis. {$6164} Thus, in at least some embodiments, non—human animals as bed, herein provide improved animal models for inflammatory and/or autoimmune diseases, disorders or conditions and can he used for the development and/or identification of therapeutic agents for the treatment, prevention and/or inhibiting one or more phenotypes (or symptoms) of an inflammatory and/or autoimmune disease, disorder or condition. in some ments, an inflammatory and/or autoimmune e, disorder or condition is present in one or more organs or tissues of a non-human animal described herein. in some certain ments, one or more organs or tissues es spleen, liver, lymph nodes, kidney, bone marrow, and h1ood. {39165} Non—human animals as described herein also provide an in viva system for identifying a therapeutic agent for treating, preventing and/or inhibiting one or more symptoms of an inflammatory and/or autoimmune disease, disorder or condition, in some embodiments, an inhibitory effect ot‘a therapeutic agent is determined in viva, by administering said eutic agent to a non—human animal that has a C90RF72 disruption as described herein, and develops an inflammatory and/or autoimmune disease, disorder or ion alter 8 weeks ot‘age. In various embodiments, an inflammatory and/or autoimmune e, disorder or ion is or comprises glomerulonephritis or hepatitis. {auras} Non-human animals may be administered a therapeutic agent to be tested by any convenient route, for example by systemic injection, pumps for long~term exposure, or direct intracerebral injection. Such animals may be included in a behavior study, so as to determine the effect of the therapeutic agent on the behavior, e.g., motor behavior, of the nonhuman s compared to appropriate control nonvliurnan animals that did not receive the therapeutic agent. A biopsy or anatomical evaluation of animal spinal cord, muscle and/or brain tissue may also be perforated, and/or a sample of blood or C3? may be collected. {$6167} Non—human animals as bed herein provide an improved in viva system and source of biological materials tag, cells) that lack expression of CQORFZ’Z that are useful for a variety of assays. in various embodiments, man s described herein are used to develop therapeutics that treat, prevent and/or inhibit one or more symptoms associated with a lack of CQGRPC’Z expression and/or activity, in various embodiments, non~human animals bed herein are used to identify, screen and/or develop candidate therapeutics {e.g., antibodies, siRNA, etc») that bind C90RF72, in various embodiments, non—human animals described herein are used to screen and develop candidate therapeutics (e.g., antibodies, siRNA, etc.) that block activity of C90RF72, in various embodiments, non-human animals described herein are used to determine the binding profile of antagonists and/or agonists of a ’Z polypeptide (or transcript) of a non—human animal as described herein, in some embodiments, non~ human animals described herein are used to determine the e or epitopes of one or more candidate therapeutic antibodies that bind '2. {sores} in various embodiments, non—human animals described herein are used to determine the pharmacokinetic profiles of a drug targeting C9GRF72. in s embodiments, one or more non—human animals described herein and one or more control or reference nonrliurnan animals are each exposed to one or more eandidate drugs targeting C9ORF72 at various doses (egt, (ll mgr/leg, 02 mg/kg, 0,3 mgl’lrg, 0.4 nag/kg, 0.5 mg/lrg, 1 mg/kg, 2 mtg/leg, 3 org/kg, 4 mg/kg, 5 mtg/mg, 7.5 org/kg, ltl mpg/kg, l5 mg/lige 2t) rag/kg, 25 mg/kg, 30 ring/lege kg, or 50 mg/kg or more), Candidate eutic antibodies may be dosed via any desired route ot‘adrninistration including parenteral and non—parenteral routes of administration. Parenteral routes include, e.g., intravenous, rterial, intraportal, intramuscular, subcutaneous, intraperitoneal, intrasninal, hecal, intracerebroventricular, intracranial, intrapleural or other routes of injection, renteral routes e, age, oral, nasal, transdermal, pulmonary, rectal, , vaginal, ocular. Administration may also be by continuous on, local administration, sustained release from implants (gels, nes or the like), and/or intravenous injection. Blood is isolated from non—human animals (humanized and control) at various time points (e.g.,, 0 hr, 6 hr, l day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, l0 days, ll days, or up to 30 or more days). Various assays may be performed to determine the pharmacokinetic profiles of administered drugs targeting 2 using samples obtained from non—human animals as described herein including, but not d to, total lgG, anti—therapeutic antibody response, agglutination, etc. {99169} in s embodiments, non-human animals as described herein are used to measure the therapeutic effect of blocking, modulating, and/or inhibiting C90RF72 activity (or C90Rl‘72 signaling, or CQORWZ—mediated interactions) and the effect on gene expression as a result of cellular changes. in various embodiments, a non—human animal as described herein or cells isolated therefrom are d to a drug targeting C90RF72 ofthe nonnhuman animal and, after a subsequent period of time, analyzed for effects on C90RF72—dependent processes (or interactions), for example, endosomal trafficking, immune homeostasis, or motor neuron and/or non~motor neuron function. [littl'llll Cells iron: non—human animals as described herein can be isolated and used on an ad hoc basis, or can be maintained in culture for many generations. in s embodiments, cells from a non-human animal as described herein are immortalized (eg, via use ot‘a virus) and maintained in culture indefinitely (eg, in serial cultures). {$6171} in various embodiments, cells and/or non~human animals as described herein are used in s immunization regimens to determine the C9ORF'72—mediated functions in the immune response to an antigen (cg, a B cell response). in some embodiments, ate therapeutics that bind, or block one or more functions of, C90Rf‘72 are characterized in a non—human animal described herein. Suitable measurements e various cellular assays, proliferation assays, serum innnunoglobulin analysis (eg, antibody titer), cytotoxicity assays, characterization of ligand—receptor interactions leg, irnrnunoprecipitation assays) and characterization of ligand—ligand ctions. In some embodiments, nonuhuman animals described herein are used to terize the C90RE72—mediated functions regulating an immune response to an antigen. in some embodiments, the antigen is associated with an autoimmune disease, disorder or condition. ln some ments, the antigen is ated with an inflammatory disease, disorder or condition. In some embodiments, that antigen is ated with a neurological e, disorder or condition. In some embodiments, the antigen is associated with an infectious agent (eg, a bacteri urn). in some embodiments, the antigen is a test antigen, (e.g., ovalbumin or OVA). in some embodiments, the antigen is a target associated with a disease or condition suffered by one or more human patients in need of treatment. {@172} In various embodiments, non—human animals as bed herein are used for challenge with one or more antigens to determine the therapeutic potential of compounds or biological agents to modulate C90RF72udependent regulation of an immune response, including but not limited to, the specific 8 ependent responses to a given antigen. lfil Nonvhuman animals as described herein provide an, in viva system for the analysis and testing of a drug or vaccine. in various embodiments, a candidate drug or vaccine may he delivered to one or more non—human animals described herein, tollowed by monitoring of the non,~human animals to determine one or more of the immune response to the drug or vaccine, the safety profile of the drug or e, or the effect on a disease or condition and/or one or more symptoms of a disease or condition.
Exemplary methods used to determine the safety profile include measurements of toxicity, optimal dose tration, efficacy of the drug or e, and possible risk factors Such drugs or vaccines may he improved and/or developed in such non—human animals. {@174} Vaccine efficacy may be determined in a number of ways. Briefly, non— human animals descrihed, herein are vaccinated using methods known in the art and then challenged with a vaccine or a vaccine is administered to alreadyuinfected non—human animals. The response ot‘a non—human (s) to a vaccine may he measured by monitoring of, and/or performing one or more assays on, the nonrhunian aninialts) (or cells isolated rom) to determine the efficacy of the vaccine. Tilt) response of a non" human anirnal(s) to the vaccine is then compared with control animals, using one or more measures known in the art and/or described herein. {$6175} Vaccine efficacy may fmther be determined lay viral neutralization assays. y, nonuhuman s as described herein are immunized and serum is collected on various days postnimmunization. Serial dilutions of serum are rare-incubated with a virus during which time antibodies in the serum that are ic for the virus will bind to it.
The serum mixture is then added to permissive cells to determine infectivity by a plaque assay or microneutralization assay. lf antibodies in the serum neutralize the virus, there are fewer plaques or lower relative luciferase units compared to a control group. {@9176} Non—human animals described herein provide an in viva system for assessing the pharmacolrinetic properties and/or efficacy of a drug (eg, a drug targeting C9ORE72), in various embodiments, a drug may be delivered or administered to one or more man animals as described , followed by monitoring of, or perfonning one or more assays on, the non-human animals (or cells ed therefrom) to determine the effect of the drug on the non—human animal. l’harmacokinetic properties include, but are not limited to, how an animal processes the drug into various metabolites (or detection of the presence or absence of one or more drug metabolites, including, but not limited to, toxic metabolites), drug half~life, circulating levels of drug after administration (cg, serum concentration ofdrug), anti—drug response (e.g.,, anti-drug antibodies), drug tion and distribution, route of administration, routes of excretion and/or clearance of the drug, ln some embodiments, pharmacolrinetic and codynamic properties of drugs (ego, C90RF72 modulators) are monitored in or through the use of nonuhuman animals bed herein. {3917?} ln some embodiments, performing an assay includes determining the effect on the phenotype and/or genotype of the non~human animal to which the drug is administered. ln some ments, performing an assay es determining lotnto- lot variability for a drug (e.g., a C90RP72 modulator such as, e.g., an antagonist or an agonist). In some embodiments, performing an assay includes determining the difierences between the effects of a drug administered to a non-human animal bed herein and a reference nonuhuman animal. in various embodiments, reterence non— human animals may have a cation as described herein, a modification that is ent as described herein (e.g.,, one that has a altered, disrupted, deleted, inserted, modified, etc, or otherwise non-functional C9ORF72 locus) or no modification tie, a wild type nonuhuman animal). {@9178} ary parameters that may be measured in non~human s {or in and/or using cells isolated rom) for assessing the phannacokinetic properties of a drug include, but are not limited to, agglutination, autophagy, cell division, cell death, complement—mediated hemolysis, DNA integrity, pecific antibody titer, drug metabolism, gene expression arrays, metabolic activity, mitochondrial activity, oxidative stress, ytosis, protein thesis, protein degradation, protein secretion, stress response, target tissue drug tration, nonntarget tissue drug concentration, transcriptional activity, and the like. In various embodiments, nonuhuman animals described herein are used to determine a pharmaceutically effective dose of a drug (eg, a drug targeting CQORFVZ).
EXAl‘i’iPLES fililll’l’lll The following examples are provided so as to describe to those of ordinary skill in the art how to malre and use methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Unless indicated otherwise, temperature is indicated in Celsius, and pressure is at or near atmospheric. e 1. Generation of a disruption in a non~human C90RF’?2 locus {39188} This example illustrates a targeted disruption in a $005172 locus of a rodent. in particular, this example ically describes the deletion of the entire coding sequence ota mouse C9mgf‘72 locus using a iacZ reporter construct placed in operable linkage with a mouse C§mgf72 promoter. The C907f72~lch targeting vector for creating a tion in an endogenous mouse C90ij72 locus was made as previously described (see, e.g., US. Patent No. 6,586,251; Valenzuela et al,, 2003, Nature Biotech. 2lt6):652— 659; and Adams, NC. and NW. (Sale, in Mammalian and Avian Transgenesis~--New Approaches, ed. Lois, S.P.a.C., Springer Verlag, Berlin Heidelberg, 2006). The ing d €905,672 locus is depicted in Figure lA, bottom box. 399181} Briefly, a targeting vector was ted, using bacterial artificial chromosome (BAG) clones item a mouse RP23 BAC library (Adams, DJ. et al., 2005, Genomics 86:753—758) and uced into Fl hybrid (l ZQSoSVEvl‘ac/CS 7BL6N'l'ac) embyronie stem (ES) cells followed by culturing in selection medium containing G418, Drug—resistant colonies were picked ill days after oporation and screened for correct targeting as previously described (Valenzuela et al., supra; Frendewey, D. et al., 20l 0, Methods Enzymol. 476:295—387). The VELOCIMOUSEQ?) method iara, TM. et al., 2tll 0, Methods Enzyrnol. 476285—294; ra, T.M., 2009, Methods Mol.
Biol. 530:3l b.3324; Poueymirou et al., 2007, Nat. Biotechnol. 25:9l $99) was used, in which targeted ES cells were injected into uncompacted 8—cell stage Swiss Webster embryos, to produce healthy fully ES cell "derived F0 generation mice heterozygous for the {7.905672 deletion. F0 generation heterozygous male were crossed with C57Bl6/N'Fac females to generate Fl heterozygotes that were intercrossed to produce f2 generation C9riiff72‘/‘, £790rf72w" and wild type mice for ypic analyses. A second cohort of N2F2 generation mice was generated via in vitro fertliziation (ll/F) using frozen Fl zygous sperm and oocytes from C5 7Bl6/N’l‘ac donor females. N2Fl heterozygous offspring were then intercrossed to generate NZFZ C90ijf72"/", C90}:f72+/' and wild type mice for phenotypic analysis. {@182} Phenotypic studies of F2 and N2F2 mice began at six (6) weeks of age. Mice were observed from hirth for various developmental milestones (runting, breathing, facial and limb abnormalities, skin color, posture, righting and eye opening) until 6 weeks of age, when they were housed 2—5 per cage in l2 hours of light per day at 20~ 230C, and 40—60% humidity for study. Mice were housed in 95.6 a 309.l X 133.4 mm cages (Thoren) with co‘o bedding (The Andersons Lab Bedding) and a cotton nestlet for enrichment e). In housing, the mi ie were monitored twice daily for health status and had access to normal chow (LahDiet) and water ad libitum. All animal procedures were carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory s of the National Institutes of Health. The protocol was approved by the Regeneron Pharmaceuticals lnstitutional Animal Care and Use Committee (lACUC), and all efforts were made to minimize suffering. {£39183} TAQMANKE Expression Arnalysls: ry, al and al lymph nodes, gonadal fat pad, frontal cortex, diaphragm, spinal cord, spleen and thymus tissues were dissected fresh into er stabilization reagent (QlAgen) and stored at "209C. s were homogenized, in ’l‘RlZGLthl reagent and phase separated with chloroform The aqueous phase, containing total RNA, was ed using sy Mini Kit (QlAgen) according to manufacturer's specifications. Genomic DNA was removed using MAGMAXTM M DNase Buffer and TURBQTM DNase (Amhion). mRNA was reverse~transcribed into cDNA using SUPERSCRIPTQ?) VILOTM Master Mix Script® ill RT, RNaseOUTlM recombinant ribonuclease inhibitor, proprietary helper protein, random primers, MgClZ, dNTPs; lnvitrogen by Life logies). cDNA was amplified with the TAQMAN® Gene Expression Master Mix (Applied Biosystems) using the ABl 7900HT Sequence Detection System (Applied Biosystems).
BetanActin was used as an al control gene to normalize CDNA input differences.) Thymus from wild type mice was used as a reference sample to calculate fold difference of inVRNA between samples n=5 females per tissue per genotype), Exemplary results are set forth in Figure Md. {@9184} Lan Expression Profiling: Mice were deeply anesthetized via Ketarnine/ Xylazine (120/5 trig/leg) lP injection and fixed by c ion using a 0.2% glutaraldehyde, 4% paraformaldehyde solution. Brain, ri‘oeage, lymph nodes, salivary glands, thymus, heart, lung, liver, , stomach, kidney, intestine, urogenital, muscle, and hind limb tissues were dissected, rinsed in PBS and post—fixed for 30 minutes in a 0.2% glutaraldehyde, 4% paraformaldehyde solution. Tissues were washed and incubated in X—gal ( l mg/mli) staining solution for 1—24 hours at 37%; After staining, tissues were washed, postutixed in 4% paralormaldehyde and cleared in a glycerol series of 50%, 70% and l00%. Photographs were taken with a Nikon SMZl 500 stereornieroscope and Nikon DS~Ril digital camera using NlS—Elements l) imaging Software ).
} Expression profiling was recorded at embryonic day l2.5 , 6 weeks, and 28 weeks, Representative data of the relative expression profile of li—galactosidase ([5252) in El2.5 embryos {Table 4) and 6— and 28nweel: old C90rf72‘fl mice {Table 5) are provided below (m no expression; + low expression; -+-‘+- moderate expression; +- = high expression; wt = wild type C5 "flit/6N; nd = not determined). {salsa} As shovm in Figure lB, high levels of {390,672 expression was detected in wild type (WT) gonadal fat pad, frontal cortex and spinal cords, with lower levels in the thymus, spleen, and lymph node. 72+”' (llet) mice had roughly hal t” the expression level of wild type (WT), as expected, and C,9orf72"l' (KO) mice had no detectable 2 expression. N0 ence in transcription levels of nearby loci MobSb, Ak045932, and lfhk among tested genotypes was observed, which ted that insertion of {an}: alone (i.e., coding sequence ablation) aftected expression of C§mgf72. [hilld'll Consistent with data shown in Figure 18, 24262 staining in 6 and 28 week 72"/‘ (KO) animals ed enzyme activity in several regions of the brain and spinal cord, as well as in spleen, testes, and kidney (Tables 4 and 5), which is consistent with other reports (Suzuki, N et alt, 20M, Nat. Neurosci. l6(12):l725—8; hoppers, M et al, 2015, Am. Neurol. 78(3)2425—38). Finther, less prominent staining in other tissues was observed. Reporter activity was more iimited, in intensity and scope in C90ij72+"” tissues, as expected for a singie iacZ repiacement allele. {@9188} Taken togethen this exampie demonstrates that marine C90ri’72 is expressed in various tissues of the nervous and immune systems Further, this e trates that at ieast in some tissues, expression increases with age ei‘the animal and directly eorreiates with neurological and immunological phenotypes deseribed in the foiiowing es (see heiow), C9mj72V' +--+-+— C9017 72"" -+--+-+- TAELIE S Spinal - +++ +++ eord HEM _ --i-- 'i"‘i""l" Riheage _ +- .i..+._+_ mb _ _+--+- .i..+._+_ LiVE‘X' _ x-|---+- 'i"‘i""l" Lung ~ ++ +++ Thymus ~ + ++ 8111661”! _ + +++ Example 2. Behavioral anaiysis of nonmhuman s having a disruption in a C9ori‘7’2 locus {$6189} This example demonstratesa among other things, that noii—iimnaii aiiimais (e.g? rodents) described herein deveiop ALSuiike symptoms sueh as? for example, decreased body weight and significant motor abnormaiities resulting from a disruption in, a rodent (egg mouse) C90rf72 locus as described in Example 1, {@9196} ypic studies ofmice having a disruption in 3. (390472 as described above were med at 8, 189. 37 (female) and 57460 weeks (maize). Body weight was WO 96185 measured on a bi-weelrly basis, and body composition was ed by uCT scan (Dynamic 60). Standard 24 scan was used to ize mass of the cervical region of the spine, All animal procedures were conducted in compliance with protocols ed by the Regeneron Pharmaceuticals institutional Animal Care and Use Committee. liltllllll Assessment of overall motor function was performed using blinded subjective scoring assays. Analysis ofmotor impairment was conducted using rotarod, open field locomotor, and, catwalh testing. Motor impairment score was measure using the system developed by the ALS‘ Therapy Development institute l, Gill A. et al., 2009, PLoS Gne 4:e6489). During catwalk testing, subjects walk across an illuminated gl ass platform while a video camera records from below. Gait d terssuch as stride pattern, individual paw swing speed, stance duration, and pressure are reported for each animal. This test is used to phenotype mice and evaluate novel chemical entities for their effect on motor performance. CatWalk XT is a system for quantitative assessment of loottalls and gait in rats and mice. it is used to evaluate the locomotor ability of rodents in almost any kind of experimental model of central s, peripheral nervous, muscular, or skeletal abnormality. llltllllzl Catt/Valle Gait Analysis: s are placed at the beginning of the runway ofNoldus CatWallr X’l‘ l0, with the open end in front of them. Mice spontaneously run to the end of the runway to attempt to . The camera records and the software of the system measures the footprints. 'l‘he footprints are analyzed for abnormalities in paw placement. {@9193} ()pen Field Test: Mice are placed in the Kinder Scientific open field system and evaluated for 60 s. The apparatus uses infrared beams and computer re to ate fine movements, X+Y ambulation, distance traveled, number of rearing events, time spent rearing, and immobility time. liltlllldl Rotorod: The rotorod test (llTC Life Science, Woodland Hills, CA) measures the latency for a mouse to fall from a rotating beam. The rotorod is set to the experimental regime that starts at l rpm and, accelerates up to l5 rpm over l80 seconds.
Then, the animals? latency to fall following the incremental regime is recorded. The average and maximum ofthe three longest durations of time that the animals stay on the beam without falling off are used to evaluate falling latency. Animals that manage to stay on the beam longer than ltlll s are deemed to be asymptomatic. liltilllfi} Upper motor neuron impairment presents as spasticity (i.e., rigidity), sed es, tremor, hradykinesia, and Babinshi signs. Lower motor neuron impairment presents as muscle weakness, wasting, clasping, curling and dragging of feet, and fasciculations. Bulbar impairment presents as difficulty swallowing, slurring and tongue iaseiculations. Table 6 sets forth the scoring methodology related to motor impairment, tremor and rigidity of animals during testing. Exemplary results are set forth in s 2A~2H. liltillld} As shown in Figures lA—ZH, £7905f72'/' mice trated ALS—like phenotypes such as, for example, decreased body weight, motor inactivity and gait ment. in particular, decreased body weight in C9mgf72”’l' miee as compared to wild type control mice began at about 30 weeks of age (Figure 23), Further, with the ion ot‘rotarod, significant motor impairment {e.g., significant weakness and colapsing of hind limbs towards lateral midline, as well as mild tremor and rigid hind limb muscles, 01) was observed for (390672": mice in all types of testing (Figures 2025) beginning at about 40 weeks of age, which indicated the onset r nad lower motor neuron pathology. Similar defects were not observed in wild type or heterozygous (£7.9mjf72i'fl') animals. {$6197} d and CatWalk gait analysis on C90Iff72‘fi mice demonstrated significantly decreased loco—motor behaviors and fewer rearing events, indicating hind limb impairment. CatWalk gait analyses revealed signs of impaired lower inter-limb coordination and reduced stride length, as well as bradyldnesia and dragging of hind limbs, These data indicated significant gait abnormalities as ed to wild type. No difference between wild type and C.9mgf72"/' mice was ed in regard to maximum time on the rotarod. As early as 36 weeks of age, 72'/' mice demonstrated significant and progressive motor s, {@9198} in another experiment, the lumbar portion of spinal cords from wild type and C9:)(f72"'/' mice (n=5, 6t) weeks old) were collected for histopathological analysis. No difference in total number of motor s in the spinal cords was observed (Figure 21).
However, the mean cell body area of C90rf72'/' motor neurons was significantly larger as compared to wild type (p<0.ll(l(ll). in particular, the motor neurons of C9riijf72‘/' mice demonstrated hypertrophic characteristics evidenced by significantly larger mean cell body area as compared to wild type (Figure 21)., Thus, these data indicated a possible onset of lower motor neuron pathology ing at 40 weeks ot‘age. 2016/034304 300199} in a similar experiment, motor abnormalities were assessed in wild type (C90rf72'm', n=== l4; ll female, 3 male) and C905f‘72"/' (n===l7; l2 temale, 5 male) starting at 32 weeks up to 60 weeks of age as percent of living s at a given week. Mice were weighed weekly and assessment of overall motor function was performed using d subjective scoring assays (as described . Weekly or hi—monthly clinical neurological exams were performed on the two groups ofmice looking at their motor impairment, tremor and rigidity of their hind limb muscles. For motor impairment, we followed a blinded neurological scoring scale {described ahove) from of zero (no symptoms) to tour (mouse cannot right themselves within 30 seconds ct‘being placed on their side). For tremor and rigidity, we created a scoring system with a scale from zero (no symptoms) to three (severe). All data reported as mean i SEM. Representative s are set tiirtli in Figure Zl. {00200} Locomotor behaviors were evaluated for 60 minutes every other week using the automated Gpen Field system (Kinder Scientific), rotarod test (Rota Rod, ll'l'C Life Science, Woodland Hills, CA.) and gait analysis (CatWalls: X'l‘ l0, Noldus) as described above. All data reported as mean it SEM. Representative results are set forth in Figure {00201} Using the scoring scales described ahove, the inventors observed that at around 40 weeks cfage, the C90rf72_,- mice started showing significant weakness and collapsing of their hind legs towards lateral midline, as well as mild tremor and rigid hind limb muscles (P < ), suggesting the onset of upper and lower motor neuron pathology. Further, all of the wildntype mice lived, past 60 weeks of age, but only ~53% C9mf72"/" mice (9 of l 7; 5 female, 4 male) were alive at 60 weeks of age (Figure 23, top left). Beginning at around 36 weeks of age, C905f72'”/' mice ceased gaining body weight, in contrast with the cohort of wild—type mice. {00202} From the open field assay, the ors observed that 72"’/' mice display cantly decreased locomotor behaviors compared to their wild~type counterparts (P = 0.0008). These mice also displayed significantly less rearing behaviors (P = 0.0009), which ted impairment of their hind limhs. No significant change in the maximum time mice would stay on a ng beam at any time during the study was observed n wild type and CQOWXIL mice. From. the h gait analysis, the inventors observed that (3905:7241 mice have significantly impaired lower interlimb coordination {P 0.0005) and stride length (P 0.00l3), as well as hradykinesia and dragging of hind limhs.) These data indicated significant gait alities in C90rj72"; mice as compared to wild type. Thus? the example demonstrates that, starting at around 36 weeks of age, £7907j72"/' mice show significant and progressive motor deficits as compared to wild type, {@9283} in another experiment, heterozygous {C9rJiff72-V') and homozygous (C’Qorffi' ) mice were examined using a grip strength test. Briefly, the grip strength measures the neuromuseular fimetion as maximal muscle strength of forelimha and is assessed by the grasping applied by a mouse on a grid that is connected to a sensor, ’l‘hree trials were carried out in succession ing forelimb—strength only. All grip strength values obtained were normalized against mouse body . lhe grip strength test was performed thirteen wild~type3 seven C’Qorfl2+/' and eighteen C90Iff72'/" mice at 20 weeks of age (before the onset ofmotor symptoms) and on twelve wildutypeg four C9wyf72w‘, and thirteen C9mgf72”’l' mi ee at till weeks of age. Representative data is set forth in Figure 2L. 3992364} As shown in Figure 2L, heterozygous (C90igf72+’/") mice did not show any significant motor impairment? tremor or rigidity at 60 weeks. Further, heterozygous (C9orf72l‘fl') mice did not show any ehange in grip strength at 60 weeks as compared to wild type.
} Taken together; this example demonstrates that nonuhuman animals bed above demonstrate a eahle neurodegenerative phenotype and, therefore, provide useful models of ophic lateral sclerosis (ALS) and/or frontotemporal dementia (ETD)? which man animals have a genome comprising a deletion of the entire coding ce (i.e., exons Z-lG) of an endogenous C9mf72 locus resulting from insertion of a reporter gene (e.g., Zan). Such animal models provide a useful in viva system for the development and ng of therapeutic candidates for the treatment of ALS and/or FTD.
TABLE 6 (l l 2 3 Motor impairment no phenotype clasping elapsing dz; dragging paralysis Tremor none mild moderate severe Rigidity none mild moderate severe Example 3. lmmunophenotypic analysis of nonuhnman animals having a disruption in a Cymfffl locus {@9296} This e demonstrates that non—human animals made according to Example 1 demonstrate an logical phenotype characterized by, in some embodiments, splenoinegaly and lymphadenopathy resulting from infiltration of various immune cell populations. Further, this example specifically demonsrates such non— human animals develop gloinerulcnephritis characterised by infiltration of immune cell populations in the kidney. Without wishing to he hound by any particular theory, the present ors propose that the CQORF72 locus product plays a critical role in immune lunction and the loss of CQORF’P’IZ polypeptide in non—human animals described herein is not the prominent mechanism ofALS and/or ETD disease. Various tissues were harvested from C9my‘72'f" and wild type mice for analysis (n====4_6 animals per genotype at 3, l8, and 37 weeks for females, and 9—H), l8, and 5760 weeks for . lil'll Cell preparation and flow cytometry analysis: Maximum blood volume was collected into ED’l‘A coated tubes by cardiac puncture immediately following CG; euthanization and approximately 200 at was transferred into heparinucoated tubes for FACS preparation, Spleen, hone , and cervical lymph nodes were harvested, dissociated into single cell suspensions in Dolliecco‘s 1X PBS with 2% fetal bovine serum (Stem Cell Technologies) plus 2 mM EDTA (Ambion) and filtered using s known in the art. Red blood cell (REC) lysis was performed on blood, spleen, and bone marrow using REC lysis buffer (eBioscience) or ACK Lysing Buffer (Life logies). Lymph node, spleen, and bone marrow cells were counted using a Cellometer Auto T4 Cell Viability Counter lorn Bioscience) and plated for approximately l0 n cells per well for spleen, and l million cells per well or maximum volume for lymph nodes and hone marrow, Blood was plated at maximum volume {approximately 250 pL) per well. Cells were treated with LIVE/DEAD e Aqua stain, (Life Technologies) at room temperature, spun down, and re-suspended in blocking solution {purified anti~mouse CDlo/C'DBZ mAh, Bl) Phanriingen, l :100 in EACS ) for l5 minutes on ice. Cells were stained with conjugated antibodies for minutes on ice, washed, fixed (ED Cytofin/cytopeiin kit) and washed again. Cells were finally resuspended in FACS buffer (llulhecco’s lX 'PBS with 2% fetal bovine serum, Stem Cell Technologies, plus 2 mlvl EDTA, Amhion) and analyzed on a BB 2016/034304 EACSCanto Flow Cytonieter ll or LSRFortessa Flow eter (BD enees).
FoXpS staining {eBioscience) was performed according to manufacturer's specifications. {@9298} Plasma cell staining panel: CD1 lb (Ml 1’70; Biolegend), CDl lc (Nu/ll 8; Biolegend), CD3 (l45n2Cll; Biolegend), 3220 (RAE—632; Biolegend), CDl9 (lD3; BD Pharrningen), CDl38 ; BD Pharrningen), and CD45 {304:1 l; BD Pharmingen).
M'yeloid cell staining panel: Fit/80 (EMS; Biolegend), CDl l5 (AFSQB; eBioseience), l_.-yfiG (RBo—SCS; eBioscience), CDl lb (Ml/70; eBioscience), CD45 (BO—Fl 1; Bl) Biosciences), and LytiC (AL—Zl; BD Biosoiences), dies to CD8, CD25, CD62L, CD69, CDl27, PDl (RPMl—Stl), Nlipdo were obtained from BioLegend (San Diego, CA). Foxp3 antibody was obtained from eBioscience (San Diego, CA). CD49b antibody was obtained from BD Bioseienoes (San Jose, CA). Data were analyzed, using Flowlo Software (Tree Star). Cotints for perent positive and total cell number were performed for spleen, cervical lymph nodes, bone marrow, and kidney of 30—35 weel: old females (wild type: 11:4; €3.9qu2” n=4) on a Nexelcom Bioseience Cellometer Auto 2000 Cell Viability Counter with AO/Pl Viability dye (acridine orange and propidium iodide).
Cell counts were used to determine absolute number ofcell populations observed by surface staining and graphed accordingly. {$6289} Histology: Tissues were harvested into 4% paraforinaldehyde (PEA, Electron Microscopy Sciences) or collected following transoardial perlitsion with 50 mL saline solution, 50 rnli 4% PFA in acetate buffer at plitlfi and finally 5t) rnL 4% PFA solution in borate butler at pll95. Spinal cords were collected into l5% ed by % sucrose solution in horate buffer until they d. All other tissues were post— fixed in 4% PFA and transferred to 70% ethanol after 24 or 48 hours. Paraffin embedding, sectioning, and benratoxylin and eosin (l-lc‘iaE) staining were performed by a commercial histology laboratory serv, Ina; Gerniantown, MD). lrnniunohistocheniistry for lgh/l, lgG, complement factor C3, CD45R, CD3, CDl38, and F4/8ll was completed by a commercial laboratory (Histotox Labs; r, CO). Motor neuron cell count and cell body area. were quantified using image (3‘. Motor neuron count represents the e from three slides per , n=5 mice and cell body area represents the average from three slides per animal, ill motor neurons per slide, n===5 mice. Complement Factor C3 lHC was quantified using Halon {$6218} Hematology Assays: Blood s were collected Via retro~orbital eye bleeds under isollurane anesthesia or by cardiac puncture after euthanasia by CO; inhalation in acccrdance with Regeneron LACUC protocol.) Complete Blood Count (CBC) with dillerential was peribrrned en 20 uL cf whole bleed using l-lemavet 950 (Drew Seientitic Group) and clinical chemistry was run an serum, samples using ,AlB'VlA 1800 Chemistry System (Siemans Medical Selutions USA} ELISAs were performed on plasma samples using the tellewing: Mouse lgG toid Factor ELlSA Kit and Mouse lgh/l Rhettrnatcid Factcr ELISA Kit (Shihayagi C0,, Ltd), Mcuse sDNA Tetal lg FililSA kit, Mouse Anti,~Nuclear dies (ANA) 'l‘otal lg FllSA litita Meuse AntimSm (Smith n) ’l’ctal lg ELlSA lritg Mouse ardielipin 'l‘etal lg ELISA kit (Alpha Diagnostic 1nd,), and lgG and lgM mouse ELISA kit {Aheant} according to manufacturer‘s specificaticns. Samples were read en a arnax M5 Microplate Reader at 450 um (Molecular Devices)» Samples were analyzed in duplicate and averaged for mean values lFN "y, ll; l 0,, £er23 an4, life; 114.40, lL— l 2 total, iL— l 7,, M67949 and TNF~CX were measured in plasma s using a Multi~Spot® ill—plea electrechernilurninescence ion assay (Mesa Scale Discovery) acccrding to manufacturers specificatiens and read an a Mess Sector S 600 plate reader at 620 nm (Meso Scale Discovery), Samples were analyzed in duplicate and averaged for mean value. [002i ll RNA lsulatitnt.3 Sequencing and is: Spleen and al lymph ncdes were dissected fresh into RNALater stabilizatien reagent (Qiagen) and stared at QGDC.
Tetal RNA was isolated using MagMAXTM Nucleic Acid lselation Kit (Amman) per manufacturer‘s specificaticns. RNA was quantified using UV spectrupheterneter and RNA integrity was evaluated by Qiaxcel (Qiagen), PeiyA mRNA was purified from total RNA using Dynabeads nilLNA kit (lnvitregen) and strand specific RNA—Seq libraries were prepared with the ScriptSeq RNA—seq Library Preparatien kit (lllurnina).
RNA—Seq libraries were sequenced in a length ct” 33bp using q 2000 NOS sequencer (llluniina). Gene expression levels were derived frern raw sequencing reads using Nimbusz, an RNAnSeq re developed by Regeneron Pharmaceuticals lnc.
E00212} Urinalysis Method; Urine samples were obtained via spet ccllectien and urinary albumin eencentraticn was determined with Alhuwell M indirect ccrnpetitive ELlSA kit (Execellg elphia, PA) Urinary nine can" centratien was assayed using the Creatinine Cempanicn l<it (Exocell). Assays were performed according in manul‘acturer’s instructions and data obtained were used te calculate the urine alburnin~ tevcreatinine ratin (ACE), lllllllfil Statistical analysis: Statistical and cal analyses were performed using Graphl’ad Prism re on 3.0). Data were analysed using Student’s unpaired t— test and onevway analysis of variance (ANOVA) Results were considered statistically significant at p values 42.05 {error bars depict s.e.m.). Exemplary results are set forth in Figures 3Au3AL. {near 4} As shown in Figures EA—ED, C907f72'fl mice developed significant enlargement of the spleen as compared to wild type and C90igf72+’/' mice, Further, cervical lymph nodes were progressively larger with age (Figures 3An3D), Thus, as early as 8 weeks of age, £79057?” mice demonstrate enlarged spleens and cervical lymph nodes. Such enlargements were palpable in the cervical regions of all C907f72$ mice, hut not wild type or CQOKWBH‘ mic-e. Upon r investigation, such masses were palpable by l2 weeks of age in female C’Qrtif72'/" mice; and in both male and female C9:)(f72"'/' mice by l8 weeks. Upon dissection, the masses proved to originate from cervical lymph nodes, with enlargement observed as early as 8 weeks of age (Figure 3A).
A full dissection also revealed, additional enlarged lymph nodes hout the body, most notably mesenteric lymph nodes in older 2'f" mice (>35 weeks). Peyer’s patches were also notably enlarged and splenomegaly was apparent in C90rf72'fl' mice by 8 weeks of age (figure 3B, bottom left), Only nine of l7 C90rf72$ mice lived past 60 weeks of age, whereas all wild type mice suhj ected to periodic neurological timction tests survived to the end of the mental period. At about l 8—24 weeks of age, splenornegaly and cervical lymph node hyperplasia were well—established in all {Wolf/72" ”I” mice and £79urf72"/" body weight curves began to n as compared to wild type and, C90if72H— mice (cg, Figure EB). liltiZlS} CBC data with differential of whole blood shows that C9067?" mice develop a significant increase in circulating neutrophils, eoslnophils and monocytes as compared to wild type, while demonstrating a significant decrease in circulating lymphocytes (Figure 3E). CBC data from C905f72"’l' mice (cg, 34-38 weeks old) also demonstrated that circulating white blood cell differential was d as compared to wild type mice.
The inventors observed, that the significant increase in monocytes and neutrophlls, and a decrease in cytes in C9my‘72'/' mice as compared to wild type mice were detectable as early as 8 weeks of age. {$6216} ll&E ng revealed a mixed, population of cells with multiple morphologies in the spleen and al lymph nodes of C905f‘72"’/‘ mice (Figures 3F, 2016/034304 3G), Specifically, cervical lymph nodes viewed at 4x power showed an abundance of large round cells characterized by variably distinct cell borders and with moderate to abundant eosinophilic characteristics (figure 3F). Occasionally, foamy cytoplasm was observed in the expanded cell population, When viewed at 60x power, cells within cervical lymph nodes demonstrated plasmacytoid morphology (blue arrows) intermixed with phils (yellow arrows) and other mature lymphocytes (Figure 3G). Cells that appeared to be consistent with macrophages (green ) were also t as were Mott cells (intermittently, red arrow; abnormal plasma cells with condensed immunoglobulins), all ofwhich indicated c inflammation in cervical lymph nodes of C9073f7241' mice (Figure 3G). {$6217} The observed enlargement of spleens and lymph nodes in C90rf‘72'/" mice ted a neoplastic or immune dysregulation e process, which has not been usly reported in ALS—FTD patients, Histopathological analysis on C9ffl§f72$ lymphoid tissues (i.e., ng lymph node and spleen sections from 860 week old mice with hematoxylin and eosin (H&E)) ed that the basic cellular organization of enlarged lymph nodes was ved. Further, ll—lC staining confirmed the presence of B cells (CD43?) in the cortex and T cells (CD3+) between the follicles and in the paracortex zone, However, there was expansion of the cortical and medullary nodal architecture by a cell population consisting mostly of large round cells with variably distinct borders, and with a single round nucleus surrounded by eosinophilic and foamy cytoplasm» A similar cellular infiltrate was also present in the spleen, predominantly located within the red pulp, which expanded the splenic architecture and, as a result, splenic weights in C,9mf72"/' mice. it was also noted that an abundance of plasmacytoid cells containing perin uclear halos, tent with plasma cell morphology, along with occasional Mott cells were t. Similar mixed infiltrates were not observed in wild type and C9030?“ (heterozygous) mice. [mill 8} The large, round cell tion did not stain consistently with CD45R, CD3, or €3,138, but was ly positive for Pal/’80, a macrophage lineage marker, lllC signal was predominant on the cell membrane but scant in the cytoplasm due to the highly vacuolated cytoplasm. in contrast, WT and heterozygous control F4/80 staining was characteristic of cytoplasmic and membranous staining pattern expected for macrophages, with overall Ell/’80 signal more intense than that observed in C’90ij’72"/‘ mice (see below, e.g., Figure 3F). lllll Heidi and lHC es of additional organs from mice aged 8—60 weelrs revealed sporadic thymus rnedullary hyperplasia and bone marrow local lihrosis and/or myeloid hyperplasia in, certain C,9m;f72"’/' mice. A more common observation was the presence of a prominent population of dendritic cells found in the liver and kidneys of null mice. These elongated to angular cells were Flt/titl'l and logically resemhled typical dendritic cells (DC), though larger in size and more us. rlhey were more pronounced in £7.9mjf72"/‘ liver as early as 8 weeks compared with wild typea though there was no evidence of associated liver disease. We also observed an increase in Ell/80+ cells in C9olgf‘72'l" kidney at 8 weeks that became more prominent with age. DC were located primarily within the outer medulla, where they formed aggregates in the vicinity of the macula densa and nt tubules, along with ent cuffs around glomeruli in association with lymphocytes. We noted increasing infiltrates of mixed leukocytes in the kidney as mice aged, accompanied by varying degrees of immune—mediated glomerular disease that was well—established by 3560 weeks of age. No evidence of inflannnation in brain or spinal cord tissue was observed in any animals examined. "thus, the spleen and lymph nodes were the major sites of immune pathology in C9orj'72' mice with indications of secondary progressive glomerular disease in the kidney, {$6228} As shown in Figure 3H, C90zf72$ male mice demonstrate increasing numbers of (fill lb'CDl 'BZZtl'lCDl 9+ B cells in the cervical lymph nodes, while showing comparable or decreased percentages of these same B cells in spleen, bone marrow and blood as compared to wild type. B cells that are transitioning to plasma cells (BZZGIBWOWCD l 9mid’li‘w“) and mature plasma cells (BZZZOEOW’I‘CD l Slim" CD45+CDl find/ii) appeared to increase with age in the , al lymph nodes, and hone marrow of (399572"; male mice as compared to wild type (Figure 3t). {$6221} The tage ofB cells {CD45+CDl§+) was either unchanged or reduced in female C,90rf72"/' mice as compared to wild type, depending on the organ examined (e.g., cervical lymph nodes). C907f72'fl female mice demonstrate increasing percentages of B cells transitioning to plasma cells/plasma blasts +Cl)lQimBZZGmCll138+) and mature plasma cells (CD45+CD19'3220'CDl38+) in spleen, lymph node and hone marrow as compared to wild type (Figures 3}). We did not e any tent differences between £790rf72”l‘ and control mice in these cell types in the hlood. 'l‘aken together, these data demonstrated an advancing adaptive immune response in C90rf72‘/' mice. 3992322} As shown in Figures 3K and BL, increasing percentages of neutrophils (CDl lhl‘LytSG‘lLytiCl) were observed in the spleen ofmale and female C90rgf72"/‘ mice as they aged, increases were also observed in the cervical lymph nodes of C,9mj72"’l' male mice between 9~l 8 weeks and, C90rf72‘fl female mice at all time points examined.
Granulocyte populations were also increased in bone marrow and blood with varying cance at most time points. inflammatory monocytes (CDl lhl, CDl lSl', LytiGW“, l..y6Chlgh) were significantly increased in £7.9mjf72"/‘ mice as compared to wild type for spleen, cervical lymph nodes, bone marrow and blood, during at least one time point of testing (Figures 3K and 3L, middle row). Similar increases of nt tes (CDl lhl‘CDl l5lLyéGlow’l”Ly6tf5mi‘y") over time was also observed in C9mj72”’l' mice in spleen, bone marrow and blood, with decreases noted in the cervical lymph nodes {Figures 3K and EL, bottom row, tively). As shown in Figure 3M, sed populations of Flt/8t)+ macrophages in the spleen, cervical lymph nodes, kidney and hone marrow were observed in €90,672"; mice as compared to wild type, llltl2323l Histopatholcgical analysis in the context of (IDA-fill, CD3 and CD138 expression was also analyzed in the spleen and cervical lymph nodes of wild type and C905f72'fl' mice (Figures 3N, .30), Sections were viewed at 4x and 60x power. hi the spleen, 7241 mice demonstrated a loss of normal follicular morphology (Figure 3N). The white pulp areas were enlarged and stie with ill—defined borders. lation of cells with abundant light pink cytoplasm (plasmadike cells) was observed CDl 38 staining did not differ ly from wild type mice and some of the proliferating cells in the center of the white pulp did not stain with CD45R, CD3, or CD l 38. The spleens of wild type mice demonstrated essentially normal morphology with white pulp areas composed of central T cells (anti—CD3 ll-lC) surrounded by a rim ofB cells {anti—CD45R lllC), and, CDl38 staining for plasma cells was minimal (Figure 3N, left). {99224} in the cervical lymph nodes, (39027724; mice demonstrated islands of lymphoid tissues scattered amongst large aggregates ofround cells with a single nucleus and abundant eosinophilic cytoplasm (Figure 30). These cells ed the normal architecture, but vely normal B cell and T cell areas remained {evident in the center ofCD3 and CD45R stained sections). The abnormal cells intermittently stained with CD3, CD138 (arrow in bottom right image of Figure 30), and CDélfill but were generally negative for all three markers. Wild type mice showed normal lymph node morphology (Figure 30, right) CD45R iinmunostaining (B cell) was found at the periphery surrounding T cell (CD3) zones and (fill 38 rarely stained cells in the medulla. {£39225} Histopathological analysis in the context of F4/80 sion was also analyzed in the spleen and cervical lymph nodes of wild type and C90rf72‘fl mice (Figure 3F). Sections were viewed at 4x and 60x power. The data demonstrated ve F41"St} staining (macrophages) in C9067?” mice, which ates with the large foamy cell infiltrate observed in H&F staining {described shove), Extracellular Flt/80 ng was also observed, in the red pulp of spleen in wow?" mice. + cell number increased with age item 868 weeks in 72"/' mice, and was increased in ‘72'f" lymph nodes as compared to wild type lymph nodes. {$6226} ”l‘otal (7945+ (common leukocyte antigen) cell counts were increased in all tissues examined trorn C,9025f72"/' mice, which was consistent with immune infiltration ed. However, CDAlS+ percentages compared to total cell populations assayed were either ged or d as compared to wild type (Figure 33). Specific antibody panels were employed to determine if homeostasis within leukocyte subsets was altered, Neutrophil (CDélSl'CDl lblLyfiillLyoCimCDl l5') and total inonocyte (CD45'l‘Clle lb+CDl 15+) percentages were variably increased in lymph node, spleen, and bone marrow of (390:7:72"; mice as compared to wild type (Figure 3K and BL). increase in Fit/80+ macrophages (CD45'lCDl lh'l'Fll/litlllnyotl') was additionally observed in the spleen, lymph node, kidney and blood (Figure 3M). interestingly, although more cells stained positively with Fit/80 in tissues from CQOWZWL mice, the overall signal was less intense than that observed in wild type mice, which indicates a more widespread but less concentrated F4/8ll ll-lC profile (Figure 3?). Lde and LydC staining revealed an increased percentage of inflammatory monocytes (CD45lCDl lhl'CDl lSl‘l,y'6G'l,y'6Chi) in spleen, lymph node, , and blood of C90fi72‘fl mice. {@9227} Additional FACS is was done on 3065 week old females, a time point of specific interest as the majority of null mice have developed renal pathology but remained viable. As shown in Figure 3Q, total cell counts performed on whole tissue demonstrated a significant increase in absolute cell counts by flow cytoinetry for various compartments in {yam/724' mice. The identity of such increases was determined using flow cytometry employing various markers for myeloid dendritic cells, NE. cells, and T cells (Figures 3an3AC). Myeloid dendritic cells (CD45+CD1lb+CDllc+MllCll+) were increased by percent and total cell count in €90rf72"’/' mice as compared to wild type s the NK cell (NKp46+CTD49h+) fraction was decreased (Figure 3R). t CDZlS+ (leukocyte common antigen; stains all white blood cells) cells is comparable between wild type and C90ijf72"/' mice tissues, however, total cell counts are significantly increased, which indicates a significant infiltration of immune cells (Figure BS). Staining with T cell—specific markers CD4+ {helper T cell population) and CD8+ (cytotoxic T cell population) demonstrated decreased percentages of T cell tions (Figures 3T6AC). As shown by THC, lar profiling and CBC s, decreases observed, in lymphocyte populations may reflect the increase in proportion ofmyeloid cells. {@228} As shown above, percentages of (fiDdS’lCDBJF and (fiDdS'l‘Cl34+ cells were overall reduced in C90igf72'fl mice as compared to wild type mice, which was lilrely a consequence of the increase in tion olmyelcid cells consistent with gene signature data. Tn constrast, total cell counts for these T cell populations were increased in TL mice. This was reflective of the gross expansion of lymphoid tissue and overt immune ation observed. CD8+ and CD4+ T cell populations were further subdivided based on the expression of additional surfa re activation markers (Figures 3V" 3AC). For Cl)? T cells, a significantly increased percentage of the early activation and eftector memory T cell markers C369 and CD44, respectively was observed in C90rj‘72‘ ’L mice as ed to wild type (Figures 3V and 3X). Further, an increased percentage ofT cells expressing FEM a co—inhibitory receptor that is upregulated on activated cells and plays an important role in down—regulating the immune , was observed in C9orjf72"/' mice as compared to wild type (Figure 32). Cervical lymph nodes demonstrated increased expression of CD44 and PD—l although C969 expression was decreased (Figures SV—EAA). For €134+ T cells, significant increases in the percentage of (7944, and Pill in spleen, lymph nodes, kidney and blood were observed in C90rf72'/' mice as compared to wild type, with values comparable to wild type in the bone marrow es 3U, 3W, 3AA). t CD69 expression was increased in spleen, cervical lymph nodes and kidney with varying cance e 3Y3. Concomitant with the increase in activated T cells, increased percentages ot‘Cllll»+l*”or§l}I*‘s+ regulatory T cells in spleens and lymph node was observed in C90472‘L mice as ccmpared to wild type (Figure 3AB). Also, the splenic compartment demonstrated a reduced expression of CD62L and CD127 (Figure 3AC), which are are expressed on naive or central memory T cells and are down regulated once T cells become activated. Cell count measurements also demonstrated significant increases in C90472'fi mice with varying significance. {@9229} Data from cytokine panels es 3A3, 3AE and 3AF) demonstrated elevated cytokines in the serum of 868 week old C90rf‘72": mice. in particular}, at 18 weeks ofage, levels of anl'i, lL—lll, 'l‘l‘ano. and lL—lZZ (total) were significantly increased in the serum of male C'9orgf72"/' mice as compared to wild type (Figure BAD).
For these same cytokines, significant elevated levels were observed in C90rf72'/' mice as compared to (T901772 H" mice. These data indicated systemic activation of macrophages in these mice. in all male mice ed (8—58 weeks old)? £7907j72"’/' mice trate a significant increase in circulating levels of lFN—y, lL—lll, l'Lle (total)? ll_.—l 7' and "ENE—ct as compared to wild type mice. ln 8—38 week old female C9mgf72”’l' mice, a significant increase in circulating levels of lL—l 0, lL— l2 (total), lli~l79 Tbilisi and MCP—l as well as an increasing trend for lFN—v, was observed as compared to wild type mice. lL-l 2 (total) was increased approximately 6~fold in €90,772"; mice as ed to wild type mice. anlG, lL—l7a, and ’l’NF—d were also elevated. although to a lesser extent. No changes in the levels MIL-l ii, ill—2, or ill—4 were observed, and while there was increased lL—o in some C90igf72'/' mice as ed to wild type? this difference did not reach significance. Levels of the chemolrine MCPul were significantly increased in , but not male C.9m;f72"’l' mice (Figures 3AE and SAP), and lliNry was significantly increased in males with some females trating a slight increase (Figures BAE and 3Al'). ’l’husg overall increased levels of pro—inflammatory cytokines are observed as early as 8 weelrs in C9tirf72‘/' mice with varying significance as compared to wild type mice. 3992336} As shown in Figures 3A6n3Al‘ig. aging C905f72‘fl mice develop increased severity of glomerulonephritis. This result was ed in HELE staining and Ell/till ll-lC in liver and kidney (Figure EAK). For example, increased Flt/80 staining by ll-lC on 8 week old C90rf72‘fi liver trated increased infiltration of macrophages, while large FAWN macrophage cell infiltration was observed in the kidney of 38—week old female mice e SAK). sed blood urea nitrogen by serum chemistry correlated with kidney disease in f7241'mice, while increased serum globulin content ted an inflammatory condition. Normal blood urea nitrogen in mice ranges from 8—33 mg/dL, while globulin levels normally range from l~4 g/dL (see, eg, Zaiasj J. et al., 2009,. 3. Am. Assoc. Lab. Animal Sci. 48(4):387—39l3). fititlilfill Further is of lridneys revealed large, Emil/80+ clear cells that had characteristics ot‘dendritic cells present in high numbers ed around the glomeruli 01335—41 week old C902ff72-7L mice e SAK). Mild to moderate degrees of ulonephritis in kidneys of 35430 weelrmold 2"" mice was observed by tidal? staining {Figure SAK). in more ly affected s, glorneruli were enlarged hypereellular, and showed mesangeal proliferation and leukocytic infiltration.
Manifestation of immtme—mediated disease was e with observed thickening of the capillary walls and proliferation of the parietal epithelium in some glomerulh while others showed expansion of the mesangium with an acellularg eosinophilic hyaline material, consistent with glomerulosclercsis and a variable degree of periglornerular fibrosis, interestingly? these areas did not prove positive for aniyloid deposition upon Congo red staining. Tubular changes included cortical and medullary tubular dilatation and the presence of hyaline proteinaceous casts and tubular ilia with degeneration/regeneration. Such changes were not observed in wild type mice. A serum chemistry panel revealing elevated blood urea nitrogen legs} Figure 3AG) and decreased serum albumin was consistent with impaired glomerular filtration that correlated to histological renal findinvs in C90r 2'” mice as corn ared to wild ‘ e mice» :3 P {@9232} To further measure the severilty of kidney disease observed in null mice, H&E stained lridney sections were blindly scored for membranoproliferative glomerulonephritisg interstitial mononuclear mation, hyaline cast formation, glomerulosclerosise and basophilic tubules; categories of renal disease associated with immune ed glornerulonephropathyo As shown in Figure 9A, weighted graphs of histopathological scoring s demonstrate that the most significant renal changes observed in null mice are associated with membranoprolilerative glomerulonephritis. individual histopath scores are represented {Figure 93) to show that all null mice display minimal to severe i'anoproliferative glornerulonephritis with occasional evidence of additional disease categories in more severely attected animals. Score of , lmiininial, 2=mild, 3=moderate and 4=severe Urine ACR measurements assayed at l4 weelr (Figure 9C, top) and 24 weel: (Figure 9Cg bottom) time points from the same cohort ofmice indicate onset rninuria in C9ort72—f— mice with age. Heterozygous mice displayed values comparable to WT consistent with the absence of an observed phenotype. fitill2333l Alsc, C90igf72'/' mice demonstrated increased levels of total lgG and lglvl antcantibcdies by ELISA as compared tc wild type mice, which indicated autoimmune disease (Figure 3Al) and ecrrespcnds tc increased serum globulin levels cbserved by serum chemistry (Figure 3AG, top panel). Additionally, serum ELlSA cf C90;y’72"l‘ mice indicated significantly elevated levels of circulating double stranded DNA (dsDNA) antibedies, antinuclear antibcdies (ANA), anti-Smith (anti—Sm) antibcdies and anti—Cardiclipin antibcdies as compared to wild type mice. ANA are auteantibcdies that bind centents cf the cell nucleus, AntindsDNA anti'bcdies are a type (ifANA dy that specifically binds dcuble stranded DNA and ardiclipin antibcdies are directed against phcsphclipid ccmpcnents cf the rnitcchendrial ne. {$6234} Further, C90rf‘72"/" mice demonstrated a significant increase in circulating rheumatoid factor (RF) antibodies as early as 8 weeks cfage (Figure BAH). increased serum globulin and auteantibcdy ccntent may indicate any cne cfvarieus disease conditions, for e, bone marrow diserder, autcinimune disease, ehrcnic inflammatory ccnditienls), liver disease, kidney disease, infections, etc, To give but one example, systemic lupus erythematcsus (SLE) is characterized by high titers cf tibodies against many cell membrane and intracellular antigens. For example, anti~Sm antibodies, which are directed against core units of small nuclear r‘ibenuclecprcteins (snRNPs), are a specific marker for SLE. {@9235} increased auteantibcdy titers in lupus patients are pcsitively correlated with an sed frequency cf ating T ular helper ('l‘lh) cells (Xu, H. er al, Cell Immunol’ 295,, 46-—-5l (205)), lnterrogaticn of this ic cell populaticn (C Dd-l-CXCRS-l-CDs’lll-l-lCOEli-PDl -+-Bcl—6+-) in spleen, cervical LN, mesenteric LN, and bleed by FACS analysis revealed icantly increased Tlh cell pepulaticns in 72",_ s compared with ccntrols e l0). Elevated ’l‘lh cells were alsc Observed in C9067?” BM that did not reach significance e l6), Collectively, these cbservatiens t the ncticn that an immune respcnse similar tn human SLE Occurs in the absence cf Cgcrfle? expressicn. {eases} Expansions in plasma cells and transiticning l3 cells/plasmahlasts can be associated with specific neeplasms such as multiple myelcma and piasmacytoma, as well as auteimmune ccnditiens. The spleen and lymph node cf C9mf72'fl' mice were enlarged and an immune infiltrate was notably cbvieus, r the infiltrating cells were negative for B cell markers {CD45R}, and were Ell/80+ with characteristics of foamy macrophages. While the population of these cells was large, they ed regional areas of these tissues considered appropriate for the lineage, and did not obliterate the basic ecture of these tissues. in addition, the mitotic index was low, with only rare mitoses observed. Thus, neoplasm seemed unlikely. There was, however, a population ofplasma cells and occasional Mott cells present in these tissues, and evidence of glomerulonephritis, which indicated autoimmunity in CQOWXIL mice. As shown above, both lgG and lgM—type anti—RE titers were significantly elevated in C90rf72‘/" mice as compared to wild type and C9w3f72l"/‘ (heterozygous) mice (Figure BAH). Further, total serum levels ot‘lgG and lglvl were icantly elevated in C9073f724l mice starting at 8 weeks of age (Figures 3AG and 3A1) which was consistent with serum chemistry panels showing elevated globulin in 2'/' mice. {99237} Autoantibodies to glomerular basement membrane, or tion of soluble immune complexes within the ular capillaries, followed by complement fixation and inflammation has been ed to cause renal e (immunemmediated glomernlonephritis). To determine if the observed increase in total immunoglobulin and autoantibody levels were contributing to glomerulonephritis, lHC was med on kidney sections from 8—63 week old 2'fl and wild type mice for total lgG and lgM {Figure SAL). As described herein, C90rf72"/' mice demonstrate clear evidence of glomenilonephritis at both the serum and histological level. in particular, kidneys from Cilorfia ”’L mice trated increased lgG immunostaining as compared to wild type mice at all time points examined. Further, at 8 weeks, C9orjf72"/' kidneys showed difiiise, intense lllC signal in the vasculatnre and tubular epithelium of the medulla and cortex. Correlating with the onset of glomerulonephritis pathology, a marked increase in glornerular lgG and lgM staining was observed by 38 weeks. Staining for both lgG and lgM was also frequently associated with the parietal layer of Bowman's capsule.
Staining for lgG was occasionally observed in the urinary space and/or within al renal tubules, ting impaired glomerular filtration firnction (i.e., leak that exceeds resorptive ty). intense lgG staining was present in tubule epithelial cells in s with severe disease, consistent with reabsorption of abundant lgG. Similar yet less intense staining was observed for lglVl less frequently. Staining in tic glorneruli was diminished as compared to wild type, which was consistent with impaired blood flow to these units (i.e., vascular loops had been replaced with matrix or mesangial cells). However, glomeruli that retained clear vascular loops tended to have sed lgG as compared to wild type. Fine granular deposits and/or linear staining of lgG and lgM associated with ar membranes was frequently observed under high magnification and suggests immune x depositiont {@9238} Complement factor C3 deposition is commonly associated with immunoglobulin deposits on nt membranes in the kidney. ll-lC for complement factor C3 revealed increased staining in ular tufts of CQOWZJ- mice as compared to wild type (Figure SAL), Granular and linear staining was most prominent on the membranes of the visceral layer of ular capsule, prominently delineating the capillary loops and podocytes. {$6239} Gene signature data from molecular profiling in spleen and cervical lymph nodes (ll—l 0 and 35 week wild type and C9047?” mice) indicated infiltration of hage, monocyte, and granulocyte cell populations. Depletion of T 8; B cells was also observed, which may reflect the increase in proportion ofmyeloid cells present.
Global chical analyses primarily separated brain samples by gender and age, rather than genotype, indicating that profiling diiferences in this tissue were due to the basic biology of the samples and genotype. Only C905f72 expression was consistently different in brain tissue across both ages and genders. in contrast, s from spleen and lymph node clustered based on genotype, with age and sex only secondary, which indicated that transcriptome differences in these organs were the result of changes in C90ijf72 expression, Further, over lGG loci associated with immune function trated significant expression differences in £79orf72"/' mice as compared to wild type for both males and females at early and late time points. Spleen and lymph node gene signatures in C9023f72'/' mice indicated myeloid infiltration with a simultaneous se in the lymphocytic footprint consistent with CBC data (see above) demonstrating able total leukocyte counts among strains due to a balance between elevated myeloid cells and decreased lymphocytes. in a comparison of biosets, the strongest profiling matches were to immune response signatures, mouse models of s inflammatory conditions, and human ious diseases As shown in this example? iinmunophenotyping data demonstrated that mice having a disruption in a (3190672 locus (C901f72‘fi) develop megaly and lymphadenopathy as early as 8 weeks of age.) ln particular, CBC data showed an increase in circulating monocytest neutrophils and eosinophils in Cf)orf72"’/' mice as well as decreased lymphocytes in the blood heginning at 8 weeks. Also, cervical lymph nodes get ssively larger with age in male {58 weeks) and temale (37 weeks) 2'/' mice. This example also specifically demonstrates that {Turf/72%" mice develop ulonephritis (i,,e., infiltration ot‘F4/80+ macrophages in the kidney) and autoimmune disease (i.e., significant elevated levels lglvl and lgG autoantibodies) as they age. Thus, this example specifically described that rodents having a disruption in a (T902772 locus as described in Example l trate detectable ahnormalities in the periphery and circulation as early as about 8 weeks of age, ln particular, (390672 ablation lead to a chronic systemic immune se resulting in ed inflammatory cytokines and myeloid expansion in several compartments.
Example Kl. Adnunistratlon of neurotoxins to non-human animals having a disruption in a C90rf72 locus [hill/ill} This experiment demonstrates that administration otvarious toxins to non" human animals described herein can exacerbate aspects of the observed ALS~lil<e phenotype, lit particular, this example specifically demonstrates that administration of various toxins to €905,672" mice mildly exacerbates the ALSnlike motor phenotype and increases oxidative stress on motor neurons, but does not aiTect the increased inactivity and gait abnormalities in these mice, This example also trates that motor neurons of C90rf72'/' mice develop significant mitochondrial dysfunction.) [hill/ill Briefly, mouse embryonic stem cells are cultured and differentiated into motor neurons, during an eight—day period. The first day, previously frozen mouse embryonic stem cells are thawed and added to a l5 ml falcon tube with live trill of embryonic stem cell medium (ES medium: DMEM with l5% PBS, l% pen/strep, 1% glutamine, l% non—essential amino acids, l% nucleosides, 0.l% li—mercaptoethanol, 1% ~pyruvate, and hit” at 10000 unit/mL). The tube is then centrifuged, for live minutes at 800 rpm. The supernatant is aspirated and the tells suspended in l0 mL ot‘ES medium. The cells are then plated on a T75 ilasl< that is coated with l0 mL of 0. l% gelatin and incubated for 30 s at 37°C- to facilitate ment to the flask .
The cells are then incubated overnight. The following day the medium exchanged with fresh medium for survival. {eases} The ing day medium is aspirated from the flash The flash is washed with ll) mL of PBS, and, then 5 mL of trypsin is added to detach the cells from the WO 96185 bottom of the flash. The cells are incubated for five minutes at 37%. Detachment is confirmed by checking the flash: under a cope. Differentiation medium (ll) niL DFNK : 414% advanced DMEM/Flz 44% neurohasal, l% repta l% glutamirrd 0.le B~mercaptoethanol, l0% knock-out serum, replacement) is added to the flash in order to stop the trypsin reaction. The solution from the flash is collected into a falcon tube and eentrifirged for five s at 800 rpm. The supernatant is aspirated and the cells suspended in l2 mL DFNK medium. The cells are then plated in cell culture dishes and put in an incubator overnight at 37°C. The following day, solution with the cells is transferred to a falcon tuhe and fuged for two s at 5th rpm.
The atant is aspirated and the cells suspended in l2 rnL DFNK. medium. The cells are then plated in new cell culture dishes and put in an incubator oveniight. The next day, medium from the dish is collected and transferred to a falcon tube. The tube is then centrifuged for two minutes at Still rpm and then the supernatant aspirated. The cells are suspended in 36 mL DFNK mediuing with retinoic acid at l old and ened antagonist at 0.25 tilt/l final concentration, for motor neuron dit‘terentiation. The medium is split to 12 ml. per dish across three dishes. liltildfi} After three days ernhryoid hodies (EB) that form are dissociated. First, BBS are collected and transferred to a falcon tube. The cells are then centrifuged for two minutes at 590 rpm and the atant is aspirated. The cells are then washed with 4 mL oi‘PBSnglucose. Next, 4 mL trypsin is added to the cells for chemical dissociation and are incubated for five minutes. Then, l mL of horse serum is added to stop the trypsin reaction. The EBs settle for five minutes and the supernatant is aspirated. 2 mL of PBS—glucose—BNase is added and the cells are mechanically dissociated ten times.
The cells settle for 5 min and dissociated, cells are transferred to a separate tube. 2 rnL of PBS~glucoseuDNase is added to non—dissociated cells and the mechanical dissociation is repeated. The cells settle for live minutes and then dissociated cells are transferred to a separate tuhe. 'l‘he dissociated cells are centrifuged for five minutes at 800 rpm and then the supernatant is aspirated. The dissociated cells are suspended in 5 rnL of embryonic“ stem cell motor neuron medium (ESMN medium: neurohasal, 2% 327, 2% horse serum, l% pen ./'strept., 0.25% glutamine, 0.0l% E3~rnercaptoethanoh l0 rig/nil; BENT, l0 ng/mL CNTF, l0 ng/rnL GDNF). The cells are centrifuged for five minutes at 300 rpm.
The supernatant is aspirated and the cells are suspended in ESMN medium. The cells are then counted using a Countess automatic cell counter (Life Technologies) and 0.5 to l n cells are plated with 2 mL ESMN medium per well in each six well plate. 'l‘he sells remain in either ESMN (control) or ESMN with BMAA at {ll—ilk} ul‘vl concentrations. Cell counts were taken using the with {3.4%trypan hlue using a mean cell diameter setting of >20 um (motor neurons). Exemplary results are set forth in, Figure 4, {@244} In another experiment, wild type miee were administered weekly hp. inj eetions of BMAA (560 rug/kg) or PBS (control) for six weeks starting at week l (Figure 5A, top). Body weight measurements were recorded each week up to 6 weeks, week l4 and week l8. is efmeter ment via retarod, open field locomoter, and catwalk testing (as described above) were recorded at the start of the ment (week 0, lG—weeks old), every other week up te 6 weeks, week l4 and week 18.
Exemplary results are set forth in Figures SAuSD. {@245} For wild type mice, the data demonstrated that BMAA kills cultured wild type motor neurons in a desevdependent manner via AMPA/kainite receptor—mediated pathway. Further, weekly injections tip.) of BMAA did not induce an ALS~like phenotype in wild type mice (Figures SA-fiD). Similar s were ed when using lllll g BMAA. ltltl24el in another experiment, aged (he, 32nweek old) wild type and C9erjf72"/' mice were administered weekly in. injections of BMAA (Still mg/kg) or PBS (control) for six weeks. Body weight measurements were recorded each week up to '33 weeks starting at day zero (i.e., 32 weeks). Analysis ofmeter impairment via rotarod, open field loeomoter, and, catwalk testing (as descrihed above) were also recorded at the start ef the experiment (week i}, llluweeks old) and every other week up to 38 weeks. Table 6 sets forth the scoring methodology related to motor impairment, tremor and rigidity of animals during testing. ary results are set forth in Figures (ilk—(SE. The data demonstrated that administration of BMAA to C9my'F72‘/' mice mildly exacerbates the ke meter phenotype, but does net affect the increased vity and gait abnormalities of these mice. {66247} in another experiment, meter neurons from C90;fli"72.l- miee were cultured as described above (see also Figure 7) and treated with antisense eligenueleotides that selectively target sense strand repeat—containing RNAs and reduce sense-eriented RNA feel witheut affecting overall C9023??? expression. Treatment was fellowed by addition of let} rnM BMAA. Survival and oxidative stress of cultured motor neurons were measured at days l and 7. Briefly, oxidative stress of plated embryenie stem cell— WO 96185 2016/034304 derived motor neurons ibed, above) was assessed by measuring the Reactive ()xygen Species (ROS) levels in the cells using Life Technologies' CellROX Qxidative Stress Green reagent at a final tration of 5 aid and incubating for 30 minutes at 370C. After tione cells were washed three times with PBS and fluorescence was measured using a standard microplate fluorometry. ary results are set torth in Figure 7. The data demonstrated that exposure of CmeF72"’/' motor neurons to BMAA causes increased ive stress.
E00248} in another experimentg mitochondrial on was ined in wild type and Cv90rf72‘fl mice. Brieflyg the ratio of mitochondrial to nuclear DNA of embryonic stem cell—derived motor neurons (described above) was measured by DNA isolation using DNAzol reagent rogen). Purity and ty of DNA were assessed using Nanodrop 2000 spechtropliotometer (Thermo Scientific) and NoanUANT mouse mitochondrial to nuclear ratio kit (Novagen) according to manufacturers specifications.
Seahorse Bioscience XFe96 Analyzer was utilized to assess mitochondrial respiration of embryonic stem cell—derived motor neurons. Percent oxygen consumption rate to the first measurement of wild type mice was recorded for l2 measurements using the XFe96 Extracellular Flux Analyzer. The mean of first three measurements represented basal respiration, the next three after addition ofoligomycin (l ) represented proton leak? the ence between basal respiration and proton leak represented ATP production, the next three measurements alter the addition ot‘FCCP (l ph/l) represented maximal respiration, the difference between maximal and basal respiration represented spare respiratory ty and the final three after the addition of rotenone/antimycin A (0.5 uM) represented non—mitochondrial respiration. All data were collected from at least three independent experiments and are reported as mean :1: SEM. Student’s t-test was performed for statistical analysis comparing values of wild type mice to C90rf72-/- mice with * for P 5 0.012 ** for P S 0.0L and *** for P 5 0.001. Exemplary results are set forth in Figure 3. {00249} Previous reports have demonstrated that ATP depletion results in intracellular accumulation ot‘l‘la+, leading to pathological cellular hypertrophy (Liang D. et alw 2007, Neurosurg. Focus 22(5):E2). As shown in Figure 83 using motor neurons differentiated from stern cells of wild type and (1905(724- mice (described above; see also Wichterle H. et alw 2002, Cell ll0t3):385n97), C90Iff72‘fl mice demonstrated a failure in the Nanlsi ATPase pump due to lack ofATP and/or compromise of the cell membrane. in constrast, no difference in survival and oxidatiye stress was observed in wild type or C90rf72"/' neurons (Figure 8, top). Interestingly, a greater amount of mitochondrial to nuclear DNA was ohserved in motor neurons from C90;;f72"/' mice (Figure 8, top right), as well as a significantly lower (P < ltll} mitochondrial respiration rate as compared to wild type motor neurons {Figure 8, bottom left). Further, hasal respiration, ATP production, maximal respiration, proton leak and spare respiratory capacity were all significantly lower in 72l; motor neruons as compared to wild type (Figure 8, lower right). Thus, motor neurons from C905f724’" mice demonstrate significant ondrial dysfunction that likely leads to cellular damage and hypertrophy.
EtlllZSlll The t example specifically demonstrates that C907f72'fl mice show AliS—lilre motor deficits. Further, this example highlights that while BMAA kills motor neurons in an AMPA/kainate—mediated glutamate excitotoxicity y, exposure to BMAA is not enough to induce e in viva. er, exposure to BMAA only mildly exacerbates the ALS~disease phenotype in C'9oWe"’/' mice, Therefore, the data presented herein suggest that, at least in some ments, the loss of C9orl72 protein in C90lj‘72v/l" mice is not the prominent mechanism oi‘ALS—FTD disease. {@9251} Taken er, the present sure ically demonstrates that CQQWZ" mice made according to Example 1 demonstrate complete ablation of the C90rf72 locus. Further, as described herein, C9mgf7J’A mice develop several ct phenotypes throughout development characterized by, for example, significant motor deficits and a disruption in immune system and mitochondrial function. For example, C'Qorff72‘f' mice develop an autoimmune phenotype characterized by a significant increase in serum autoantihody tration and infiltration of various immune cells into the spleen, lymph nodes, hone marrow, kidney and blood. lnterestingly, immtmophenotyping data described herein illustrate that C9orjf72 gene product plays a critical role in immune system homeostasis and neuronal health. In particular, splenomegaly and lymphadenopathy in C90rf72'fl' mice are a result of ration of a number of cell populations including plasma cells, monocytes, granulocytes, and most notably, F4/8ll+ macrophages as early as 8 weeks of age and ssive h 60 weeks of age.
Cytokine panel and molecular profiling data strongly suggest an increased Th l .I’l‘vlacrophage activating pathway in (390672"; mice. Thus, the present disclosure specifically demonstrates that haploinsuftioiency is unlikely the main cause ofALS —E'l‘l:l ogy in the context of C9ffi’f72 and provides a novel role for C90rf72 in immune function and homeostasis in a comprehensive phenotypic analysis of a non—human animal with global C9mgf72 ablation.
EQUIVALENTS {@9252} Having thus descrihed several aspects of at least one ernhodirnent of this invention, it is to he appreciated by those skilled in the art that various alterations, modifications, and improvements will readily occur to those skilled in the art. Such alterations, modifications, and improvements are ed to be part of this disclosure, and are intended to be within the spirit and scope of the invention, Accordingly, the foregoing description and drawing are by way of e only and the invention is described in detail by the claims that follow.
} Use of ordinal terms such as ”first," "second," "third," etc.., in the claims to modify a claim element does not hy itself connote any priority, ence, or order of one claim element over r or the temporal order in which acts of a method are performed, but are used merely as labels to distinguish one claim element having a n name from another element having a same name (hut for use of the ordinal term) to distinguish the claim elements. {@254} The articles "a" and "an" in the specification and in the , unless clearly indicated to the contrary, should he understood to include the plural referents. Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one, or all of the group s are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes emhodiments in which exactly one member of the group is t in, ed in, or otherwise relevant to a given t or process. The ion also includes emhodiments in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process. Furthermore, it is to he understood that the invention encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, descriptive terms, etc, from one or more of the listed claims is introduced into another claim dependent on the same base claim (or, as relevant, any other claim) unless otherwise indicated or unless it would he t to one of ordinary skill in the art that a contradiction or inconsistency would arise. Where elements are presented as lists, (e.g.,, in Marhush group or similar format) it is to be 2016/034304 understood, that each subgroup of the elements is also disclosed, and any eleinent(s) can be removed from the group. it should be understood that, in general, Where the invention, or aspects of the invention, is/‘are referred to as comprising particular tal features? etc-.9 certain embodiments of the invention or aspects of the invention consist, or consist essentially of, such elements, features, etc. For es of simplicity those embodiments have not in every case been specifically set forth in so many words herein. lt should also be understood that any embodiment or aspect of the invention can be explicitly excluded from the claimsa less of whether the specilic exclusion is recited in the specification. {@255} Those skilled in the art will appreciate typical rds of deviation or error attributable to values obtained in assays or other processes described herein. {@9256} The publications, websites and other reterence materials referenced herein to be the background of the invention and to provide onal detail regarding its practice are hereby incorporated by reference.

Claims (9)

The claims defining the invention are as follows:
1. A rodent comprising in its genome a deletion of the coding portion of exon 2 through the coding portion of exon 11 of an endogenous C9orf72 locus, wherein the rodent develops one or more symptoms of (i) immune system dysregulation or dysfunction and/or (ii) motor and neurological abnormalities similar to those found in human motor neuron diseases.
2. The rodent of claim 1, wherein the C9orf72 locus ses a er gene.
3. The rodent of claim 2, wherein the reporter gene is operably linked to a C9orf72 promoter, optionally wherein the C9orf72 promoter is an endogenous promoter.
4. The rodent of either claim 2 or claim 3, wherein the reporter gene is operably linked to exon 1 of the C9orf72 locus.
5. The rodent of any one of claims 2-4, wherein the reporter gene is selected from the group consisting of lacZ, luciferase, green fluorescent protein (GFP), enhanced GFP (eGFP), cyan fluorescent protein (CFP), yellow fluorescent protein (YFP), enhanced yellow fluorescent protein (eYFP), blue fluorescent protein (BFP), enhanced blue fluorescent protein , DsRed, and MmGFP.
6. The rodent of any one of the preceding claims, wherein the rodent is a rat or a mouse.
7. The rodent of any one of the preceding claims, wherein the rodent develops one or more symptoms of ophic Lateral Sclerosis (ALS) and/or temporal Dementia (FTD) during development, optionally after about 36 weeks of age or after about 40 weeks of age.
8. The rodent of any one of the preceding claims, n the rodent develops progressive motor deficits after about 36 weeks of age, develops lower motor neuron pathology after about 40 weeks of age, and/or develops a decrease in body weight after about 36 weeks of age.
9. The rodent of any one of the preceding claims, wherein the rodent develops one or more of the following: (i) mitochondrial dysfunction in motor neurons, n mitochondrial ction is characterized by a decrease in one or more of mitochondrial respiration, basal respiration, maximal respiration, spare respiratory capacity, ATP production and proton leak; or characterized by an increase in the mitochondrial to nuclear DNA ratio as compared to the mitochondrial to nuclear DNA ratio of the motor neurons of a control or reference rodent; (ii) one or more symptoms of glomerulonephritis, optionally after about 35 weeks of age or after about 35-41 weeks of age inclusive; (iii) splenomegaly after about 8 weeks of age; (iv) lymphadenopathy after about 8 weeks of age, wherein optionally lymphadenopathy is palpable after about 12-18 weeks of age inclusive; (v) an infiltration of one or more of macrophages, monocytes and ocytes into the spleen, cervical lymph nodes, bone marrow and/or blood; and/or an infiltration of F
NZ737000A 2015-05-29 2016-05-26 Non-human animals having a disruption in a c9orf72 locus NZ737000B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201562168171P 2015-05-29 2015-05-29
US62/168,171 2015-05-29
US201562232658P 2015-09-25 2015-09-25
US62/232,658 2015-09-25
US201562245382P 2015-10-23 2015-10-23
US62/245,382 2015-10-23
PCT/US2016/034304 WO2016196185A1 (en) 2015-05-29 2016-05-26 Non-human animals having a disruption in a c9orf72 locus

Publications (2)

Publication Number Publication Date
NZ737000A NZ737000A (en) 2021-02-26
NZ737000B2 true NZ737000B2 (en) 2021-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
US11547101B2 (en) Non-human animals having a disruption in a C9ORF72 locus
JP7193509B2 (en) Non-human animals exhibiting attenuation of upper and lower motor neuron function and perception
KR20210005661A (en) Pathogen-resistant animals with modified aminopeptidase N (ANPEP) gene
NZ737000B2 (en) Non-human animals having a disruption in a c9orf72 locus