WO2014145776A2 - Msp and its receptors in the therapy of amyotrophic lateral sclerosis - Google Patents

Msp and its receptors in the therapy of amyotrophic lateral sclerosis Download PDF

Info

Publication number
WO2014145776A2
WO2014145776A2 PCT/US2014/030598 US2014030598W WO2014145776A2 WO 2014145776 A2 WO2014145776 A2 WO 2014145776A2 US 2014030598 W US2014030598 W US 2014030598W WO 2014145776 A2 WO2014145776 A2 WO 2014145776A2
Authority
WO
WIPO (PCT)
Prior art keywords
composition
msp
vap
protein
als
Prior art date
Application number
PCT/US2014/030598
Other languages
French (fr)
Other versions
WO2014145776A3 (en
Inventor
Hugo Bellen
Benjamin Russell ARENKIEL
Wan Hee YOON
Guang Lin
Isabella GARCIA
Hiroshi Tsuda
Michael Miller
Joanna JANKOWSKY
Original Assignee
Baylor College Of Medicine
Uab Research Foundation
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 Baylor College Of Medicine, Uab Research Foundation filed Critical Baylor College Of Medicine
Publication of WO2014145776A2 publication Critical patent/WO2014145776A2/en
Publication of WO2014145776A3 publication Critical patent/WO2014145776A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Embodiments of the disclosure are directed at least to the fields of neurobiology, molecular biology, cell biology, neuropharmacology, and medicine.
  • ALS Amyotrophic Lateral Sclerosis
  • ALS is a neurological disease that affects motor neurons in the brain and spinal cord. ALS is characterized by the progressive degeneration and death of motor neurons that control voluntary muscle activity. Reduced synaptic transmission to muscles causes initial symptoms that include twitching, cramping, muscle weakness and stiffness, slurred speech and difficulty swallowing, which gradually worsen as the disease progresses. Late stage symptoms include decreased strength and muscular control, muscular atrophy, difficulty breathing, drooling, gagging, head drop, difficulty walking, and paralysis. Most patients diagnosed with ALS die within 5 years due to cachexia (wasting syndrome) or respiratory failure.
  • Familial ALS which is responsible for 5-10 % of ALS cases is associated with mutations in several genes, including C9orf72, SOD1, TARDBP, FUS, ANG, ALS 2, SETX, and VapB genes. The majority of ALS cases are sporadic, for which no genetic association have been identified.
  • Riluzole inhibits glutamate release, inactivates voltage-dependent sodium channels and interferes with intracellular events that follow transmitter binding at excitatory amino acid receptors. Riluzole does not halt the disease process, but has been shown to delay time to death by a few months. There exists a need for improved ALS therapeutic agents that go beyond symptomatic treatment and target the underlying mechanisms of ALS.
  • Embodiments of the disclosure concern methods and compositions related to the treatment and/or prevention of one or more neurological disorders.
  • embodiments of the disclosure concern methods and compositions related to the treatment and/or prevention of one or more neurological disorders.
  • the neurological disorder may be of any kind, but in specific embodiments the neurological disorder is amyotrophic lateral sclerosis (ALS).
  • a therapeutically effective amount of one or more compounds are provided to the individual diagnosed as having a neurological disorder or suspected of having a neurological disorder or at risk for having a neurological disorder.
  • the neurological disorder is associated with impaired function of vesicle associated membrane protein (VAMP)-associated proteins (Vaps).
  • VAMP vesicle associated membrane protein
  • the neurological disorder is associated with a decrease in and/or mislocalization of VapB.
  • the neurological disorder is associated with a decrease in the secretion of the amino-terminal domain of VapB, named the MSP domain (Major Sperm Protein).
  • the neurological disorder is associated with loss of MSP.
  • delivery of Vap MSP to an individual with a neurological disorder allows for amelioration of at least one symptom of the disorder.
  • the neurological disorder is ALS that may be of any type, including sporadic or familial ALS.
  • the disclosure concerns therapy treats VAPB-related ALS or SOD1- related ALS.
  • MSP Major Sperm Protein
  • SEQ ID NO. 2 the N-terminal 125-amino acid domain of the VapB protein
  • compositions for treating ALS comprise a therapeutically effective amount of MSP.
  • a functionally active derivative of MSP is provided to the individual, such as one that has one or more alterations compared to SEQ ID NO:2; in specific embodiments, there are one, two, three, four, five, six, seven, eight, nine, or ten or more alterations in the MSP derivative compared to SEQ ID NO:2.
  • the MSP derivative lacks part of its N-terminus and/or C-terminus. In some cases, one or more internal amino acids are lacking compared to SEQ ID NO:2.
  • the MSP or MSP derivative may be provided to the individual by any suitable means, such as intravenously, orally, subcutaneously, and so forth, although in specific cases there is peripheral delivery.
  • a method for treating a patient with ALS comprising providing a therapeutically effective amount of a pharmaceutical composition of MSP to the patient.
  • the MSP is provided to the patient multiple times.
  • the ALS is familial or sporadic.
  • the individual has symptoms of or has been diagnosed with amyotrophic lateral sclerosis.
  • the MSP may be administered to the patient intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intrapleurally, intranasally, intravitreally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularly, mucosally, intrapericardially, orally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, or via a lavage, for example.
  • a pharmaceutical composition for treating ALS comprising a therapeutically effective amount of MSP.
  • the MSP comprises or is the amino-terminal 125 amino acid domain of VapB protein.
  • the composition is a fragment, derivative, shuffling product, conjugate, or any modified product of MSP.
  • the more than the MSP domain of VAPB is utilized in methods and/or compositions of the disclosure; in some cases, most or all of VAPB is employed in the methods and/or compositions.
  • An example of VAPB nucleotide sequence is at GenBank® Accession No. AY358464; an example of VAPB amino acid sequence is at GenBank®
  • compositions for treating ALS comprising a therapeutically effective amount of a molecule that binds to, activates, or inhibits EphA4, Robo, and/or Dlar receptors (or human equivalents thereof).
  • compositions for treating ALS comprising an amino-terminal domain of VapB protein that is greater than or less than 125 amino acids.
  • compositions for treating ALS comprising any domain of VapB protein that activates, binds to, or inhibits EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • a VAPB paralog referred to as VAPA may be employed in the disclosure (see, for example, SEQ ID NO:25 as the human VAPA sequence with SEQ ID NO:26 as its respective MSP domain).
  • the VAPA protein has a MSP domain very similar to the VAPB MSP domain, and in particular embodiments the two MSP domains are functionally equivalent.
  • compositions for treating ALS comprise a therapeutically effective amount of SEQ ID NO:26 or
  • a functionally active derivative of MSP is provided to the individual, such as one that has one or more alterations compared to SEQ ID NO:26; in specific embodiments, there are one, two, three, four, five, six, seven, eight, nine, or ten or more alterations in the MSP derivative compared to SEQ ID NO:26.
  • the MSP derivative lacks part of its N-terminus and/or C-terminus. In some cases, one or more internal amino acids are lacking compared to SEQ ID NO:26.
  • a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual comprising the step of providing a therapeutically effective amount of a Major Sperm Protein (MSP) composition or functionally active fragment or derivative thereof to the individual.
  • MSP Major Sperm Protein
  • the neurological disorder is amyotrophic lateral sclerosis (ALS), including familial or sporadic.
  • the individual has at least one symptom of ALS.
  • the individual is suspected of having ALS.
  • the method further comprises the step of diagnosing ALS in the individual.
  • the MSP composition is provided to the individual multiple times.
  • the MSP composition may be administered to the individual intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intrapleurally, intranasally, intravitreally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularly, mucosally, intrapericardially, orally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, or via a lavage.
  • the composition is provided in proteinaceous form, although it may be provided in nucleic acid form (including in an expression vector, such as a viral vector of any kind).
  • the individual is provided an additional therapy for the neurological disorder or therapy for at least one symptom thereof.
  • the neurological disorder is ALS.
  • the additional therapy is riluzole.
  • compositions for treating ALS comprising MSP or functionally active fragment or derivative thereof in a pharmaceutically acceptable excipient.
  • kits comprising a pharmaceutical composition as disclosed herein, said composition housed in a suitable container.
  • the kit further comprises an additional therapy for a neurological disorder or therapy for at least one symptom thereof.
  • a pharmaceutical composition comprising a MSP composition or functionally active fragment or derivative thereof.
  • the composition comprises one or more modifications.
  • the one or more modifications extend the half life of the composition.
  • the MSP composition comprises one or more polyethylene glycol groups; one or more immunoglobulins; at least one D amino acid; and/or a label, tag, or both.
  • the MSP composition is fused in-frame with another polypeptide.
  • the functionally active fragment or derivative thereof is at least 80%, 85%, 90%, 95%, 97%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26.
  • the functionally active fragment or derivative thereof comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid alterations compared to SEQ ID NO:2 or SEQ ID NO:26.
  • An alteration may comprise an amino acid substitution, deletion, addition, or inversion.
  • a functionally active fragment or derivative thereof is no more than 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40 35, 30, or 25 amino acids in length.
  • a functionally active fragment or derivative thereof comprises at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 120 contiguous amino acids of SEQ ID NO:2 or SEQ ID NO:26.
  • the functionally active fragment or derivative thereof comprises a N-terminal truncation.
  • a N- terminal truncation may comprise absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
  • a functionally active fragment or derivative thereof comprises a C-terminal truncation, such as a C-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
  • a functionally active fragment or derivative thereof comprises a N-terminal truncation and a C-terminal truncation.
  • a MSP composition comprises an antibody fragment, such as Fc.
  • a MSP composition comprises collagen or albumin or both.
  • a MSP composition is further defined as a MSP multimer or a MSP dimer or a MSP trimer or a MSP oligomer.
  • a MSP composition is further defined as a peptide shuffled MSP molecule.
  • a MSP composition comprises a MSP multimer comprsing at least two, three, four, or more MSP monomers.
  • a composition comprises a collagen triple helix structure with MSP such that there is a trimer of MSP.
  • MSP is utilized in a composition with a Fc as a monomer or dimer.
  • a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of FOXOl in the individual.
  • the agent is a small molecule, nucleic acid, peptide, or protein.
  • a nucleic acid may be a polynucleotide that encodes at least part of FOXOl.
  • a method further comprises the step of providing a therapeutically effective amount of a MSP composition or functionally active fragment or derivative thereof to the individual.
  • a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of Osbp in the individual.
  • the agent is a small molecule, nucleic acid, peptide, or protein.
  • a nucleic acid may be a polynucleotide that encodes at least part of Osbp.
  • a method further comprises the step of providing a
  • Other neurological disorders that may be treated with methods and/or compositions of the disclosure include at least Alzheimer's disease; autism; cerebral palsy, dyslexia; Huntington's; multiple sclerosis; Parkinson's disease; and so forth.
  • FIG. 1A is a western blot of His-MSP eluates from Nickel affinity chromatography.
  • FIG. IB is a western blot of His-MSP before and after dialysis.
  • FIG. 1C is a western blot of His-MSP after incubation at 37 0 C for the indicated periods of time.
  • FIG. 2 displays MSP as a domain of VapB, and as a free peptide after cleavage from VapB, where it binds to EphA4, Robo, and Dlar receptors.
  • FIG. 3 shows localization of VapB to the ER and cell membrane.
  • FIG. 4 shows that P58S mutant protein accumulates in cytoplasmic inclusions.
  • FIG. 5 shows where the MSP domain is cleaved from the full length VapB protein.
  • FIG. 6A is a graph that shows that injection of His-MSP prolongs the survival of SODl G93A transgenic mice over control mice (treated with PBS).
  • FIG. 6B is a graph that shows the average survival days of His-MSP-treated SODl G93A transgenic mice was longer than control mice (treated with PBS).
  • FIG. 7 shows that injection of His-MSP rescues the motor function of SOD1 G93A transgenic mice in a rotarod assay.
  • FIG. 8 shows that fat levels in body wall muscle of wild- type and vpr-1 mutant worms.
  • A DIC images of muscle in live adult hermaphrodites. Arrowheads indicate lipid-like droplets. Bar, 5 ⁇ .
  • B Transmission electron micrographs of body wall muscle cytoplasm in wild-type and vpr-l(tml411 ) mutant hermaphrodites. Light blue color demarcates muscle boundary.
  • L Lipid-like droplet. Bar, 0.5 ⁇ .
  • C Fluorescent images of muscle in live adult hermaphrodites fed Bodipy-FAs. Closeup images of boxed areas are shown below.
  • FIG. 9 provides vpr-1 mosaic analysis.
  • A Analysis of vpr-1 genetic mosaics showing the lineages of major tissues. Each circle indicates one genetic mosaic worm. Points at which the genomic copy of vpr-1 (+) was lost and the resulting phenotype are shown.
  • B Representative DIC images of muscle in vpr-l(tml411 ) mutant mosaic worms. Ex vpr-1 (+) indicates expression of the vpr-1 genomic locus via an extrachromosomal array. Arrowheads indicate fat droplets. Bar, 5 ⁇ .
  • FIG. 10 shows the effect of tissue-specific vpr-1 expression on fat levels.
  • A DIC images of muscle in live wild- type and vpr-1 (tml411) mutant hermaphrodites expressing wild-type VPR-1 or VPR-1 (P56S) under indicated tissue-specific promoters.
  • FIG. 17 (B) Sudan Black B staining images of i mutants expressing vpr-1 under the unc-119 pan-neuronal promoter. Arrows indicate muscle fat droplets. Anterior is to the left in all panels. Wild- type controls (FIG. 17) are similar to transgenic vpr-l(tml411 ) mutants expressing uncll9p::vpr-l. Low magnification bars, 50 ⁇ ; high magnification bars, 25 ⁇ .
  • FIG. 11 demonstrates effect of Arp2/3 inactivation on muscle fat levels. DIC and fluorescent images of muscle in live 3-day-old hermaphrodite worms fed Bodipy-FAs.
  • arx-2 encodes the Arp2 component of the Arp2/3 complex. Arrowheads indicate Bodipy-FA- stained fat droplets. Bar, 5 ⁇ .
  • FIG. 12 provides DAF-16 activity in vpr-1 mutants.
  • A DIC and fluorescent images of muscle in live 3-day-old hermaphrodite worms fed Bodipy-FAs.
  • FIG. 13 shows DAF-16 localization and activity in wild- type and mutant worms.
  • A Transgenic strains expressing DAF-16::GFP under its endogenous promoter.
  • Transgenic controls raised at 20°C are similar to those raised at 20°C then shifted to 35°C for 30 minutes (see panel B for quantification). Close up images of boxed areas are shown. Anterior is to the left in all panels. Low magnification bar, 50 ⁇ ; high magnification bar, 25 ⁇ .
  • (-) incubation under normal growth condition; (+), incubation at 35°C for 30 minutes.
  • C Magnified images showing transgenic lines expressing GFP under the sod-3 promoter, arx-2 encodes Arp2. Arrows indicate vulva muscle region.
  • FIG. 14 provides the effect of DAF-16 inactivation on muscle mitochondria.
  • Oxygen consumption rate of wild- type and vpr-l(tml411 ) mutants includes published data [15] measured together with vpr- l(tml411) daf-16(mu86) mutants.
  • FIG. 15 shows the effect of DAF-16 inactivation on ATP level and lifespan.
  • A ATP concentration in wild-type and vpr-l(tml411) mutant adult extracts. *, P ⁇ 0.001 compared to wild type. Error bars represent SD. ATP concentration of wild-type and vpr- l(tml411) mutants at 1-day-old adults include published data (Han et ah, 2012) measured together with vpr-l(tml411) daf-16(mu86) mutants.
  • B Lifespan measurements of indicated genotypes. The lifespan of daf-16(mu86) mutants was similar to the wild type, as previously shown.
  • FIG. 16 demonstrates effect of Vapb ablation on fasting/refeeding energy metabolism in mice.
  • A TAG concentration in GA muscle and liver of wild-type (+/+) and Vapb knock-out (-/-) mice after 24-hour fasting (red) or 24 hours fasting followed by 6 hours of refeeding (blue).
  • B and C Quantitative RT-PCR of indicated genes in liver (B) and TA muscle (C) of wild-type (+/+) and Vapb knock-out (-/-) mice after 24-hour fasting (red) or 24 hours fasting followed by 6 hours of refeeding (blue). Relative mRNA levels are shown on the Y-axis. #, P ⁇ 0.05 compared to fed mice of the same genotype. *, P ⁇ 0.05 compared to +/+ under the same condition.
  • FIG. 17 shows Sudan Black B staining in wild type and vpr-1 mutants.
  • 1- day-old adult wild- type and vpr-1 (tml411 ) hermaphrodites were stained using Sudan Black B.
  • Arrowheads indicate fat droplets in body wall muscle. Anterior is to the left in all panels. Boxed regions are magnified 5X below. Low magnification bars, 50 ⁇ ; high magnification bars, 10 ⁇ .
  • FIG. 18 provides ER stress assays in wild-type and vpr-1 mutant worms.
  • A Integrated transgenic lines expressing GFP under the hsp-4 promoter (hsp-4p::GFP) with and without tunicamycin treatment, which induces ER stress. Anterior is to the left in all panels. Bar,
  • FIG. 19 demonstrates effect of sperm presence on muscle fat droplets in vpr-1 mutants. DIC and fluorescent images of muscle in live 3-day-old vpr-1 mutant
  • hermaphrodite worms fed Bodipy-FAs Mating with wild type (WT) males provides sperm into the uterus. Anterior is to the left in all panels. Arrowheads indicate lipid-like droplets. Bar, 50 ⁇ .
  • FIG. 20 shows loss of Vap causes accumulation of membrane proteins in the adult neurons.
  • A-B Neurons in the adult brain of 1 day-old adult flies stained with anti- Chaoptin and anti-Elav antibodies.
  • Chaoptin labels individual synapses in the lamina (dots) as previously published (Hiesinger et ah, 2005). However, it is also expressed in few cortical neurons of the brain at low levels (A, arrows).
  • Chaoptin accumulates in the cytoplasm of the neurons of the cortex (B, B' and B", arrows).
  • C-F Staining of cortical neurons in the adult brain of 1 day-old flies with anti-Robo-1 and anti-Elav antibodies (C, D) and N-Cadherin (E, F).
  • C, D cortical neurons in the adult brain of 1 day-old flies with anti-Robo-1 and anti-Elav antibodies
  • E, F N-Cadherin
  • Robo-l(D) and N-Cadherin (F) accumulates in the cytoplasm of a subset of neurons (arrows).
  • Op optic lobe.
  • FIG. 21 shows loss of Vap causes accumulation of membrane proteins in the ER of neurons.
  • A Staining of affected neurons in the adult brain of the vap null mutant. Accumulated Chaoptin protein is surrounded by BOCA, the ER marker (indicated by * in A").
  • B Staining of cortical neurons in the adult brain of 1 day-old flies expressing the membrane anchored GFP (CD8-GFP) with anti-Chaoptin and anti-GFP antibody. In the vap null mutant neurons, some neurons accumulate both CD8-GFP (arrows, B') and Chaoptin (arrows, B").
  • FIG. 22 demonstrates that loss of Vap causes an aberrant ER expansion.
  • A- D TEM analysis of neurons of control (A) and of vap null mutants (B-D). Large cytoplasmic vacuoles in the neurons of the vap null mutant (asterisk (*), B). The vacuole is contiguous with the nuclear membrane (arrow heads, C). The vacuole is also decorated with electron-dense
  • FIG. 23 demonstrates that loss of VAP causes ER stress.
  • A-D An ER stress reporter Xbpl-GFP is significantly upregulated in the neurons in the adult brain of 1 dayold the vap null mutant (B and D), but not in the control flies (A and C). A precise excision line was used as a control.
  • E-F Neurons of 1 day-old adult brain stained with anti-Bip, an ER stress marker. Bip is upregulated in the neurons of the vap null mutant (arrows in F).
  • G-H Staining of the neurons in the cortex of the adult brain of control (G) and the vap null mutant (H).
  • Ubiquitinated proteins accumulate in the cytoplasm of cortical neurons in adult vap null mutant (H and FT), but not in control adults (G and G').
  • FIG. 24 shows that Osbp colocalizes with Vap in the ER.
  • A Vap and Osbp interaction in a GST pull-down assay.
  • GST- Vap WT binds Osbp, but GST-VapALS8 binding is severely reduced.
  • GST- Vap WT and GST-VapALS8 were expressed in bacteria and purified with glutathione sepharose beads.
  • GST- Vap proteins linked to beads were incubated with lysate extracted from Schneider's S2 cells expressing Osbp. The precipitate and input were analyzed with immunoblots using anti-Osbp antibody and anti-GST antibody.
  • FIG. 25 shows that Osbp is mislocalized to the Golgi in vap null mutant cells.
  • A-B MARCM analysis shows that Osbp accumulates in cytoplasmic punctae of vap null mutant cells (B and arrows in B') but not in wild type cells (A and A').
  • C-D Osbp accumulates in the Golgi of vap null mutant cells. Immuno staining of the 1 day-old adult brain of WT
  • control C
  • vap null mutant neurons D
  • anti-Osbp C and D'
  • anti-Msl20 C" and D
  • Golgi marker Osbp barely colocalizes with the Golgi marker Ms 120 KDa
  • FIG. 26 shows that ALS8 mutation causes a partial loss of function of VAP.
  • A Comparable levels of expression of vapWT and vapALS8 transgenes. Immunoblot analysis of proteins extracted from WT (control) and transgenic flies carrying genomic vapWT (VK31) and vapALS8 (VK31) in the vap null mutant background with anti-Vap and anti-Actin antibodies.
  • B Longevity of adult flies. The vapALS8 transgene can rescue the lethality associated with loss of Vap, but VapALS8 is not as active as VapWT. An additional copy of vapWT can compensate for the defects associated with vapALS8.
  • Line F7 and M6 are P-element mediated transgenes.
  • Line VK31 is site specific integrated transgene in VK31.
  • vapALS8 transgenes rescue the lethality associated with loss of vap, but VapALS8 is not as active as VapWT.
  • Genotypes Avap; vapWT (vap null mutant carrying genomic vapWT, line Ml 1, F6 and F10).
  • vapALS8 vap null mutant carrying genomic vapALS8 , line V37, V38, V40, V42, V43, V45, and V46). All lines carry P-element mediated transgenes. Error bar represents STD. (D) Flight test. vapWT can suppress the flight defects associated with vapALS8.
  • vapALS8 (VK31) flies exhibit flight defects at day 12 (* P ⁇ 0.001 compared to control), but not at day 4 after eclosion. However, ⁇ ; vapWT/ vapALS8 flies do not show flight defects. Flies were individually dropped into a plastic cylinder, and the height at which they landed was recorded. The shorter the distance from the bottom to their landing point, the worse their ability to fly. Error bar represent SEM.
  • FIG. 27 shows that ALS8 mutation causes progressive defects in adult fly brain.
  • VapALS8 causes adult brain degeneration. Brain section of 12-day-old adult flies.
  • vapALS8 VK31
  • vapWT VK31
  • vapWT VK31
  • op optic lobe
  • cen central lobe.
  • Vap causes functional defects in motor neurons in adult flies. In the vap null mutant one day old flies, the TTM muscles are unable to follow a 200Hz stimulus but they can almost respond properly when stimulated at 10 Hz.
  • n the number of flies used. * p ⁇ 0.05 and ** p ⁇ 0.005.
  • Controls are flies that have a precise excision of P ⁇ Mae-UAS.6.11 J.
  • FIG. 28 shows that expression of human OsbpL8 suppresses the ER defects associated with loss of Vap.
  • A-F, H Immuno staining of the adult brain of WT (control) (A, B), vap null mutant (C, D), vap null mutants expressing HA-hOSBPL8 (E, F) and the vap
  • FIG. 29 demonstrates that MSP treatment induces phosphorylation of RTKs.
  • MSP stimulation activates protein phosphorylation signaling in HEK293 cell.
  • HEK293 cells were serum starved for 16hrs before stimulated with His-MSP (lOOng/ml). Cells were
  • FIG. 30 indicates that MSP treatment increases the number of primary hippocampal neuron synapses in vitro.
  • A Confocal microscopy images of MSP or PBS treated hippocampal neuron cultures. Cells are stained with the synaptic marker Bassoon, dendritic marker Map2, and nuclear marker DAPI (scale bar 50 ⁇ ).
  • FIG. 31 illustrates an example of a VAPB expression construct for utilization with recombinant adeno-associated virus 8 in an ALS model, SOD1-G93A.
  • Animals are injected intracerebroventricularly with AAV8-VAPB at birth to produce widespread expression of the viral construct in neurons throughout the brain and into the spinal cord.
  • FIG. 32 provides an example of a scheme for assessing the efficacy of VAPB treatment delivered by intracranial AAV8 injection in SOD mice .
  • Wild-type and SOD1 transgenic mice were injected with AAV8-VAPB at postnatal day 0, then tested for behavioral symptoms from postnatal day 80 until death.
  • Behavioral readouts include motor performance on the rotorod assay and an overall symptom assessment based on the scale from Vercelli et al (2008).
  • Tissue is harvested from brain, spinal cord, blood, heart, kidney, and muscle at endpoint for analysis of viral expression.
  • FIG. 33 illustrates that AAV8-VAPB treatment delays the loss of motor ability during early stages of disease.
  • SOD1 mice were administered AAV8-VAPB at birth and
  • FIG. 34 demonstrates survival rate for SOD1 transgenic mice treated with VAPB-AAV8 at birth. There is a small improvement in survival with VAPB treatment.
  • FIG. 35 shows that the initial decline in gross motor symptoms is delayed by AAV8-VAPB treatment of SOD1 transgenic mice compared to untreated SOD animals.
  • FIG. 36 demonstrates that there is strong overexpression of VAPB protein in the brains of wild- type and SOD1 transgenic mice treated at birth with AAV8-VAPB.
  • FIG. 37 demonstrates increased levels of MSP fragment in the plasma of wild-type mice treated with AAV8-VAPB at birth.
  • General embodiments provided herein concern methods and compositions for treating one or more neurological disorders, such as ALS, with part or all of the Major Sperm Protein (MSP) domain of VapB.
  • the individual that is treated shows at least one neurological symptom associated with ALS or has at least one risk factor to delay or prevent the onset of ALS.
  • MSP Major Sperm Protein
  • An exemplary risk factor is a family member with ALS and/or having one or more mutations linked to ALS.
  • ALS Amyotrophic Lateral Sclerosis
  • ALS Amyotrophic lateral sclerosis
  • Lou Gehrig's disease is a rapidly progressive, fatal neurological disease that attacks the nerve cells that control voluntary muscles, referred to motor neurons.
  • motor neurons both the upper motor neurons (in the brain) and the lower motor neurons (in the spinal cord) degenerate or die and no longer send messages to muscles, and the muscles gradually weaken and then atrophy.
  • all muscles under voluntary control are affected, and individuals lose their strength and the ability to move their arms, legs, and body.
  • Most people with ALS die from respiratory failure when muscles in the diaphragm and chest wall fail, losing the ability to breathe without a ventilator.
  • Symptoms of ALS may be overlooked initially, given their subtle nature. The earliest symptoms may include fasciculations, cramps, tight and stiff muscles (spasticity), muscle weakness affecting an arm or a leg, slurred and nasal speech, and/or difficulty chewing or swallowing. To be diagnosed with ALS, individuals need to have signs and symptoms of both upper and lower motor neuron damage that cannot be attributed to other causes. Thus, diagnosis of ALS is based on the symptoms and signs observed by a medical provider and a series of tests to rule out other diseases. Medical providers may assess over time whether symptoms such as muscle weakness, atrophy of muscles, hyperreflexia, and spasticity are increasing in their severity.
  • ALS symptoms at the onset can be similar to those of a wide variety of other, more treatable diseases or disorders, certain tests may be performed to exclude the other conditions. Such tests include electromyography (EMG), a special recording technique
  • the medical provider may utilize magnetic resonance imaging (MRI) of the brain and/or spinal cord to exclude certain conditions.
  • MRI magnetic resonance imaging
  • the physician may order tests on blood and urine samples to eliminate the possibility of other diseases as well as routine laboratory tests.
  • riluzole is provided to the individual for therapy.
  • riluzole or another ALS treatment is provided in addition to compositions of the disclosure.
  • Other medications that may be employed with the disclosure include those that alleviate certain symptoms, such as those that help reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, painkillers, depression treatment, treatment for sleep disturbances, and/or constipation medication.
  • Embodiments include the delivery of part or all of MSP to an individual in need thereof.
  • the individual may be deficient in MSP level.
  • the individual may be known to have a neurological disorder or suspected of having a neurological disorder or at risk for having a neurological disorder, such as ALS.
  • the entire MSP is provided to an individual.
  • a functionally active fragment or derivative of MSP is provided to the individual.
  • only part of the entire MSP is provided to the individual, such as one having no more than 123, 122, 121, 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, 15, or 10 amino acids of SEQ ID NO:2 or SEQ ID NO:26.
  • a particular domain of MSP is included in the MSP derivative.
  • the MSP fragment or derivative has at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 amino acids of SEQ ID NO:2 or SEQ ID NO:26.
  • the MSP derivative is modified compared to SEQ ID NO:2 or SEQ ID NO:26, such as having 1, 2, 3, 4, ,5 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more
  • the MSP derivative is at least 70%, 75%, 80%, 82%, 85%, 87%, 90%, 92%, 95%, 97%, 98%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26.
  • the MSP composition will have all or part of the amino acid sequence of SEQ ID NO:2 or SEQ ID NO:26.
  • the MSP compositions described herein may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more variant amino acids within at least, or at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106,
  • MSP or fragment or derivative thereof is delivered to an individual in proteinaceous form
  • a nucleic acid encoding the MSP or fragment or derivative thereof is delivered to the individual.
  • fragment refers to a domain of VapB that is smaller or larger than MSP that still retains the biological activity of MSP.
  • derivatives refers to compounds that are derived from chemical modifications to a starting compound.
  • shuffling product is a peptide in which an amino acid, amino acids, or groups of amino acids have been transposed from their original sequence.
  • a conjugate is molecule that has a molecule, functional group, polymer, protein or nanoparticle attached to it.
  • modified product is a molecule which has been chemically altered.
  • compositions comprising a fragment, derivative, shuffling product, conjugate or any modified product of MSP.
  • compositions for treating amyotrophic lateral sclerosis comprising a therapeutically effective amount of a molecule that binds to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • compositions for treating amyotrophic lateral sclerosis comprising a therapeutically effective amount of a molecule that activates EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • compositions for treating amyotrophic lateral sclerosis comprising a therapeutically effective amount of a molecule that inhibits EphA4, Robo, and/or Dlar receptors (and/or human
  • compositions for treating amyotrophic lateral sclerosis comprising an amino-terminal domain of VapB protein that is greater than 125 amino acids. . In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising an amino-terminal domain of VapB protein that is less than 125 amino acids. In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising any domain of VapB protein that activates to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • compositions for treating amyotrophic lateral sclerosis comprising any shuffling product of VapB protein that binds to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • compositions for treating amyotrophic lateral sclerosis comprising any shuffling product of VapB protein that inhibits EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • compositions for treating amyotrophic lateral sclerosis comprising any shuffling product of VapB protein that activates EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
  • the MSP comprises one or more modifications. Although any suitable modifications are encompassed in the disclosure, in certain embodiments the one or more modifications extend the half life of the composition.
  • the MSP composition comprises one or more polyethylene glycol groups, one or more immunoglobulins, at least one D amino acid, and/or a label, tag, or both.
  • the MSP composition is fused in-frame with another polypeptide.
  • a functionally active fragment or derivative thereof is at least 80%, 85%, 90%, 95%, 97%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26.
  • the functionally active fragment or derivative thereof may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid alterations compared to SEQ ID NO:2 or SEQ ID NO:26.
  • An alteration may be of any kind, such as an amino acid substitution, deletion, addition, or inversion.
  • a functionally active fragment or derivative thereof is no more than 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40 35, 30, or 25 amino acids in length. In certain cases, a functionally active fragment or derivative thereof comprises at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 120 contiguous amino acids of SEQ ID NO:2 or SEQ ID NO:26.
  • Particular functionally active fragments or derivatives thereof of MSP comprise a N-terminal truncation, such as a N-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
  • N-terminal truncation such as a N-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
  • Certain functionally active fragments or derivatives thereof of MSP comprise a C-terminal truncation, such as a C-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
  • Certain MSP functionally active fragments or derivatives thereof comprise a N-terminal truncation and a C-terminal truncation.
  • MSP compositions comprise an antibody, including an antibody fragment, such as Fc.
  • MSP comprises collagen or albumin.
  • Embodiments include a MSP-Fc composition, a MSP-albumin composition, MSP-collagen like scaffold molecules, and so forth, and in specific embodiments such compositions have longer pK and half-life in vivo.
  • MSP binds to different receptors.
  • a MSP composition is further defined as a MSP multimer, which may be a dimer, trimer, and so forth.
  • a MSP multimer may be comprised of 1, 2, 3, 4, 5, or more MSP monomers.
  • a MSP composition is an oligomer or a peptide shuffled MSP molecule.
  • the MSP functionally active derivative or fragment may be referred to as a biological functional equivalent, and it may comprise a polynucleotide that has been engineered to contain distinct sequences while at the same time retaining the capacity to encode the "wild- type" or standard protein. This can be accomplished to the degeneracy of the genetic code, i.e., the presence of multiple codons, which encode for the same amino acids. In one example, one of skill in the art may wish to introduce a restriction enzyme recognition sequence into a
  • polynucleotide while not disturbing the ability of that polynucleotide to encode a protein.
  • a polynucleotide may be (and encode) a biological functional equivalent with more significant changes.
  • Certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies, binding sites on substrate molecules, receptors, and such like.
  • so-called "conservative" changes do not disrupt the biological activity of the protein, as the structural change is not one that impinges of the protein's ability to carry out its designed function. However, in some cases the alteration is not a conservative substitution. It is thus contemplated by the inventors that various changes may be made in the sequence of genes and proteins disclosed herein, while still fulfilling the goals of the present disclosure.
  • polynucleotide is the concept that there is a limit to the number of changes that may be made
  • Bioly functional equivalents are thus defined herein as those proteins (and polynucleotides) in selected amino acids (or codons) may be substituted.
  • Functional activity includes the ability of the protein form to bind EphrinA4, Dlar, and/or Robo receptors (and/or human equivalents thereof).
  • MSP equivalents into the C. elegans gonad. As shown in Miller et al. 2001 and Tsuda et al. 2008, microinjecting MSPs into the gonad stimulates oocyte maturation and muscle contraction, which can be quantified.
  • Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and/or the like.
  • An analysis of the size, shape and/or type of the amino acid side-chain substituents reveals that arginine, lysine and/or histidine are all positively charged residues; that alanine, glycine and/or serine are all a similar size; and/or that phenylalanine, tryptophan and/or tyrosine all have a generally similar shape.
  • arginine, lysine and/or histidine; alanine, glycine and/or serine; and/or phenylalanine, tryptophan and/or tyrosine; are defined herein as biologically functional equivalents.
  • hydropathic index of amino acids may be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and/or charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and/or arginine (-4.5).
  • the present disclosure in many aspects, relies on the synthesis of peptides and polypeptides in cyto, via transcription and translation of appropriate polynucleotides. These peptides and polypeptides will include the twenty "natural" amino acids, and post-translational modifications thereof. However, in vitro peptide synthesis permits the use of modified and/or unusual amino acids.
  • peptide mimetics that mimic elements of protein secondary and tertiary structure are described in Johnson et al. (1993).
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and/or antigen.
  • a peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule.
  • ⁇ -turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains.
  • Beta II turns have been mimicked successfully using cyclic L-pentapeptides and those with D-amino acids.
  • Johannesson et al. (1999) report on bicyclic tripeptides with reverse turn inducing properties.
  • Beta- turns permit changed side substituents without having changes in corresponding backbone conformation, and have appropriate termini for incorporation into peptides by standard synthesis procedures.
  • Other types of mimetic turns include reverse and gamma turns. Reverse turn mimetics are disclosed in U.S. Patents 5,475,085 and 5,929,237, and gamma turn mimetics are described in U.S. Patents 5,672,681 and 5,674,976.
  • the present disclosure also encompasses a composition comprising a nucleic acid sequence encoding MSP domain as defined herein and cells harboring the nucleic acid sequence.
  • the nucleic acid molecule is a recombinant nucleic acid molecule, in particular aspects and may be synthetic. It may comprise DNA, RNA as well as PNA (peptide nucleic acid) and it may be a hybrid thereof.
  • nucleic acid molecules may contain, for example, thioester bonds and/or nucleotide analogues.
  • the modifications may be useful for the stabilization of the nucleic acid molecule against endo- and/or exonucleases in the cell.
  • the nucleic acid molecules may be transcribed by an appropriate vector comprising a chimeric gene that allows for the transcription of said nucleic acid molecule in the cell.
  • polynucleotides can be used for "gene targeting" or "gene therapeutic” approaches.
  • the nucleic acid molecules are labeled. Methods for the detection of nucleic acids are well known in the art, e.g., Southern and Northern blotting, PCR or primer extension. This embodiment may be useful for screening methods for verifying successful introduction of the nucleic acid molecules described above during gene therapy approaches.
  • the nucleic acid molecule(s) may be a recombinantly produced chimeric nucleic acid molecule comprising any of the aforementioned nucleic acid molecules either alone or in combination.
  • the nucleic acid molecule is part of a vector.
  • the present disclosure therefore also relates to a composition comprising a vector comprising the nucleic acid molecule described in the present disclosure.
  • plasmids are known to those skilled in molecular biology, the choice of which would depend on the function desired and include plasmids, cosmids, viruses, bacteriophages and other vectors used conventionally in genetic engineering. Methods that are well known to those skilled in the art can be used to construct various plasmids and vectors; see, for example, the techniques described in Sambrook et al. (1989) and Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1989), (1994). Alternatively, the polynucleotides and vectors of the disclosure can be reconstituted into liposomes for delivery to target cells. A cloning vector may be used to isolate individual sequences of DNA.
  • Typical cloning vectors include pBluescript SK, pGEM, pUC9, pBR322 and pGBT9.
  • Typical expression vectors include pTRE, pCAL-n-EK, pESP-l, pOP13CAT.
  • a vector that comprises a nucleic acid sequence that is a regulatory sequence operably linked to the nucleic acid sequence encoding MSP defined herein.
  • regulatory sequences control elements
  • the nucleic acid molecule is operatively linked to said expression control sequences allowing expression in eukaryotic or prokaryotic cells.
  • a vector is an expression vector comprising the nucleic acid molecule encoding MSP as defined herein.
  • the vector is a viral vector, such as a lentiviral vector or an adeno-associated viral vector. Lentiviral vectors are
  • control sequence refers to DNA sequences that are necessary to effect the expression of coding sequences to which they are ligated. The nature of such control sequences differs depending upon the host organism. In prokaryotes, control sequences generally include promoters, ribosomal binding sites, and terminators. In eukaryotes generally control sequences include promoters, terminators and, in some instances, enhancers, transactivators or transcription factors. The term “control sequence” is intended to include, at a minimum, all components the presence of which are necessary for expression, and may also include additional advantageous components.
  • operably linked refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" 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.
  • the control sequence is a promoter, it is obvious for a skilled person that double- stranded nucleic acid is preferably used.
  • the recited vector is an expression vector, in certain embodiments.
  • An "expression vector” is a construct that can be used to transform a selected host and provides for expression of a coding sequence in the selected host.
  • Expression vectors can for instance be cloning vectors, binary vectors or integrating vectors.
  • Expression comprises transcription of the nucleic acid molecule preferably into a translatable mRNA.
  • Regulatory elements ensuring expression in prokaryotes and/or eukaryotic cells are well known to those skilled in the art. In the case of eukaryotic cells they comprise normally promoters ensuring initiation of transcription and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript.
  • Possible regulatory elements permitting expression in prokaryotic host cells comprise, e.g., the P L , lac, trp or tac promoter in E. coli, and examples of regulatory elements permitting expression in eukaryotic host cells are the AOX1 or GAL1 promoter in yeast or the CMV-, SV40-, RSV- promoter (Rous sarcoma virus), CMV-enhancer, SV40-enhancer or a globin intron in mammalian and other animal cells.
  • Beside elements that are responsible for the initiation of transcription such regulatory elements may also comprise transcription termination signals, such as the SV40-poly- A site or the tk-poly-A site, downstream of the polynucleotide.
  • transcription termination signals such as the SV40-poly- A site or the tk-poly-A site, downstream of the polynucleotide.
  • leader sequences capable of directing the polypeptide to a cellular compartment or secreting it into the medium may be added to the coding sequence of the recited nucleic acid sequence and are well known in the art.
  • the leader sequence(s) is (are) assembled in appropriate phase with translation, initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein, or a portion thereof, into the periplasmic space or extracellular medium.
  • the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product; see supra.
  • suitable expression vectors are known in the art such as Okayama-Berg cDNA expression vector pcDVl (Pharmacia), pEF-Neo, pCDM8, pRc/CMV, pcDNAl, pcDNA3 (Invitrogen), pEF-DHFR and pEF-ADA, (Raum et al. Cancer Immunol Immunother (2001) 50(3), 141-150) or pSPORTl (GIBCO BRL).
  • the expression control sequences are eukaryotic promoter systems in vectors capable of transforming of transfecting eukaryotic host cells, but control sequences for prokaryotic hosts may also be used. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and as desired, the collection and purification of the polypeptide of the disclosure may follow. In particular embodiments, one or more encodable sequences are regulated by expression control sequences that are responsive to hypoxic environments.
  • Additional regulatory elements may include transcriptional as well as translational enhancers.
  • the above-described vectors of the disclosure comprises a selectable and/or scorable marker.
  • Selectable marker genes useful for the selection of transformed cells are well known to those skilled in the art and comprise, for example, antimetabolite resistance as the basis of selection for dhfr, which confers resistance to
  • methotrexate (Reiss, Plant Physiol. (Life-Sci. Adv.) 13 (1994), 143-149); npt, which confers
  • decarboxylase which confers resistance to the ornithine decarboxylase inhibitor, 2- (difluoromethyl)-DL-ornithine, DFMO (McConlogue, 1987, In: Current Communications in Molecular Biology, Cold Spring Harbor Laboratory ed.) or deaminase from Aspergillus terreus which confers resistance to Blasticidin S (Tamura, Biosci. Biotechnol. Biochem. 59 (1995), 2336-2338).
  • luciferase PI. Sci. 116 (1996), 59-72; Scikantha, J. Bact. 178 (1996), 121
  • green fluorescent protein a gene encoding luciferase (Giacomin, PI. Sci. 116 (1996), 59-72; Scikantha, J. Bact. 178 (1996), 121), green fluorescent protein
  • the recited nucleic acid molecule can be used in a cell, alone, or as part of a vector to express the encoded polypeptide in cells.
  • the nucleic acid molecules or vectors containing the DNA sequence(s) encoding any one of the CD 138- specific CAR constructs is introduced into the cells that in turn produce the polypeptide of interest.
  • the recited nucleic acid molecules and vectors may be designed for direct introduction or for introduction via liposomes, or viral vectors (e.g., adenoviral, retroviral) into a cell.
  • the present disclosure relates to methods to derive vectors, particularly plasmids, cosmids, viruses and bacteriophages used conventionally in genetic engineering that comprise a nucleic acid molecule encoding the polypeptide sequence of MSP defined herein.
  • said vector is an expression vector and/or a gene transfer or targeting vector.
  • Expression vectors derived from viruses such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus, may be used for delivery of the
  • nucleic acid molecules and vectors can be reconstituted into liposomes for delivery to target cells.
  • the vectors containing the nucleic acid molecules of the disclosure can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or electroporation may be used for other cellular hosts; see Sambrook, supra.
  • compositions in accordance with certain embodiments of the present disclosure comprise an effective amount of MSP or additional active ingredient dissolved or dispersed in a pharmaceutically acceptable carrier.
  • the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent.
  • the pharmaceutical composition can be introduced to a subject by any method known to those of ordinary skill in the art. Examples may include, but not be limited to administration intravenously, intradermally, intrathecally, intraarterially, intraperitoneally, intramuscularly, subcutaneously; orally, intrarectally, mucosally (intranasal, intravaginal, etc.), topically (i.e., transdermally), locally, via inhalation (e.g., aerosol inhalation), via injection, via infusion, via continuous infusion, via localized perfusion bathing target cells directly, via a
  • 40530616.1 catheter via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference).
  • lipid compositions e.g., liposomes
  • the pharmaceutical composition of the disclosure may be formulated into a composition in a free base, neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • a carrier can be a solvent or dispersion medium comprising but not limited to, water, ethanol, polyol (e.g., glycerol, propylene glycol, liquid polyethylene glycol, etc.), lipids (e.g. , triglycerides, vegetable oils, liposomes) and combinations thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin; by the maintenance of the required particle size by dispersion in carriers such as, for example liquid polyol or lipids; by the use of surfactants such as, for example hydroxypropylcellulose; or combinations thereof such methods.
  • the actual dosage amount of a composition in accordance with certain embodiments of the present disclosure administered to an animal patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event,
  • 40530616.1 determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • Methods may involve administering to the patient or subject at least or at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more doses of a therapeutic composition.
  • kits having one or more components housed in suitable container means.
  • the kit comprises an effective amount of a MSP protein or MSP fragment or derivative.
  • the components of the kits may be packaged either in aqueous media or in lyophilized form.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there are more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
  • the kits of the present disclosure also will typically include a means for containing the component(s) and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.
  • the container means may itself be a syringe, pipette, and/or other such like apparatus, from which the formulation may be applied to a desired area of the body, injected into an animal, and/or even applied to and/or mixed with the other components of the kit.
  • the components of the kit may be provided as dried powder(s).
  • the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
  • kits of the disclosure may also comprise, and/or be packaged with, an instrument for assisting with the injection/administration and/or placement of the ultimate composition within the body of an animal.
  • an instrument may be a syringe, pipette, forceps, and/or any such medically approved delivery vehicle, for example.
  • the kit comprises one or more additional agents for treatment of ALS or a symptom thereof, such as riluzole, drugs that reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, painkillers, depression treatment, treatment for sleep disturbances, and/or constipation medication, for example.
  • additional agents for treatment of ALS or a symptom thereof such as riluzole, drugs that reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, painkillers, depression treatment, treatment for sleep disturbances, and/or constipation medication, for example.
  • VAMP Vesicle-associated membrane proteins
  • VapB Vehicle-associated membrane protein-associated protein B
  • ER endoplasmic reticulum
  • MSP is the N-terminal 125 amino acid domain of VapB.
  • UPR Unfolded Protein Response
  • MSP binds to multiple growth cone guidance receptors that are present on both neurons and muscles, including the EphrinA4, Dlar, and Robo receptors.
  • MSP binding to Ephrin A4 leads to unclustering of glutamate receptors. Conversely, loss of MSP causes a hyperclustering of glutamate receptors and an increase in Ca 2+ influx, leading to neuronal excitotoxicity. Binding of MSP to Dlar and Robo is required to retain proper mitochondrial morphology, localization, and function in muscle. In the absence of MSP, muscle function is impaired. This leads to a dysfunctional motor neuron-muscle connection, resulting in a negative feed-back loop resulting from the loss of a BMP-like growth factor (GBB), which ultimately leads to neuronal loss and disease progression.
  • GBM BMP-like growth factor
  • VapB proteins do not play an important role in synaptic transmission in vivo as originally proposed (Pennetta et al. 2002). Rather the fly, worm, and mammalian VapB homologues are type II integral membrane proteins that are mostly localized to the cytoplasm and span the ER lipid bilayer (Tsuda et al. 2008; Wyles et al. 2002). They have been implicated in a variety of processes in model organisms.
  • Protein aggregates are common pathological features both in familial and sporadic form of ALS. Loss of VapB in flies causes the formation of protein aggregates and an UPR in motor neurons and certain neurons in the brain (FIG. 4). This UPR is associated with a secretory defect of transmembrane proteins from ER to plasma membrane (Yang et al. 2012; Moustaqim-Barrette et al. 2013). Interestingly, an UPR has also been shown to be associated with the mutant form of VapB found in ALS8 patients (Ince et al. 2011; Papiani et al. 2012; Qiu et al. 2013; Tsuda et al.
  • ALS as well as numerous forms of ALS including the SODl mice models and sporadic human ALS patients (Lautenschlaeger et al. 2012). Wild type SODl has been shown to be misfolded and aggregate in sporadic ALS patients carrying the Fus mutation (R521C, Pokrishevsky et al. 2012). VapB is required for ER homeostasis.
  • VapB has significant roles in lipid metabolisms and transport. Loss of VapB causes a defect in the transport of ceramide from the ER to the Golgi (Peretti et al. 2008; Perry and Ridgway 2006). Ceramide is enriched in the ER and the spinal cord of ALS patients has been reported to have twofold excess of the normal levels of ceramide (Cutler et al. 2002).
  • MSP is secreted in the fly haemolymph, the worm pseudocoelom fluid, and is present in mammalian blood and functions as a hormone (Han et al. 2012; Tsuda et al. 2008).
  • the MSP domain of VapB is evolutionarily related to a group of 28 nearly identical proteins that are abundantly expressed in C. elegans sperm (hence, the name Major Sperm Protein, MSP, Miller et al. 2001). These proteins are secreted/released from sperm by a poorly characterized mechanism (Kosinski et al.
  • MSP/EphA4 signaling contributes to the pathology of ALS.
  • Mammalian EphA4 can function as a receptor for MSP (Tsuda et al. 2008), and EphA4 has recently been implicated in ALS (Van Hoecke et al. 2012).
  • EphA4 regulates actin polymerization in muscle, which is critical to the formation/stability of the neuromuscular junctions (NMJs) (Lai et al. 2001).
  • NMJs neuromuscular junctions
  • MSP/Lar and Robo signaling also play critical roles in the pathogenesis of the disease. Two other growth cone guidance receptors expressed on muscles, Lar and Robo, function in the control of actin polymerization and depolymerization.
  • SODl transgenic mice exhibit an UPR (Tobisawa et al. 2003) and a severe loss of the VapB protein (Teuling et al. 2007). This is also observed in all sporadic ALS human patients tested to date (70 out of 70), and it is the only protein that has been shown to be absent in all loci tested that cause ALS (Teuling et al. 2007; Anagnostou et al. 2010).
  • the EphA4 receptor which is expressed on muscle in vertebrates (Lai et al. 2001), has recently been implicated in ALS (Van Hoecke et al. 2012), and EphA4 binds MSP (Tsuda et al. 2008).
  • MSP is involved in proper maintenance and function of the neuromuscular junction.
  • the cumulative effect leads to a withdrawal of the NMJs during the final stages of the disease, ultimately leading to denervation of motor neurons that is at the core of the pathology of ALS.
  • MSP By binding to its cognate muscle receptors (EphA4, Dlar, and Robo), MSP promotes proper Ca 2+ metabolism, mitochondrial dynamics, and mitochondrial function, preventing a premature negative feed-back loop to the presynaptic terminal. Therefore, MSP is used as a therapeutic agent in sporadic as well as familial ALS patients.
  • MSP 40530616.1 MSP were selected by PCR analysis using Ml 3 primers.
  • the bacmid containing His-MSP was transfected into Sf9 cells to generate baculovirus expressing His-MSP protein.
  • the lysate and bead mixture from step 4 was poured into Poly-Prep Column [Bio-Rad; cat. no. 731-1550EDU] to separate the lysate and beads by gravity at 4°C (*kept the flow-through for the determination of binding efficiency).
  • the beads were washed three times with 20ml of Wash Buffer (15 OmM NaCl, 50 mM phosphate buffer pH7.4, and 20mM imidazole) at 4 °C.
  • the His-MSP protein was eluted with 3 ml of Elution Buffer (150mM NaCl, phosphate buffer pH 7.4, and 500 mM imidazole) at 4 °C.
  • the eluted His-MSP was filtered at 4°C using an Amicon Ultra-30K device [Millipore; cat. no. UFC903024] to remove proteins greater than 30 KDa in size.
  • the His-MSP was dialyzed against 1L of 1XPBS overnight twice at 4 °C.
  • the protein concentration was measured by Bradford Assay [Bio-Rad; cat. no. 500-0006].
  • the protein purity was checked by 15% SDS-PAGE and coomassie blue staining.
  • VapB makveqvlsl epqhelkfrg pftdvvttnl klgnptdrnv 1
  • ALS is a lethal neurodegenerative disease characterized by the combined degeneration of lower and upper motor neurons (Kiernan et ah, 2011). Most ALS cases occur sporadically, although a subset of cases is inherited. These familial cases are caused by mutations in multiple genes, including in the Vapb (VAMP/synaptobrevin-associated protein B) gene.
  • Vapb Vapb
  • Vapb ALS8 that manifests as ALS or late-onset SMA, a motor neuron disease restricted to lower motor neurons
  • Vapb mutations are rare, reduced VAPB mRNA or protein levels have been reported in sporadic ALS patients, a mSODl ALS mouse model, and ALS8 patient motor neurons derived from induced pluripotent stem cells (Anagnostou et al., 2010; Teuling et al., 2007;
  • VAPB deficiency might have a function relevant in non-ALS8 patients.
  • VAPB and its paralog VAPA, are broadly expressed type II membrane proteins that are widely conserved. These VAPs have been implicated in regulating lipid transport and homeostasis, endoplasmic reticulum (ER) dynamics, and membrane trafficking (Loewen and Levine, 2005; Amarilio et al, 2005; Lev et al, 2008; Peretti et al, 2008; De Vos et al, 2012).
  • VAP vMSP is cleaved from the transmembrane domain in the cytoplasm and secreted in a cell-type specific fashion (Tsuda et al, 2008; Han et al., 2010; Han et al., 2012).
  • Secreted vMSPs antagonize Eph receptor signaling through a direct interaction with the extracellular domain (Tsuda et al., 2008).
  • the inventors have shown in C. elegans and Drosophila that neurons secrete vMSPs to regulate mitochondrial localization and function in striated muscle (Han et al., 2012).
  • vMSPs interact with muscle SAX- 3 Roundabout and CLR-1 Lar-like protein tyrosine phosphatase receptors to down-regulate CLR- 1 signaling.
  • VAP loss causes uncontrolled CLR-1 Lar-like receptor activation in body wall muscle.
  • CLR-1 stimulates actin filament assembly in the muscle belly that requires the actin- related protein 2/3 (Arp2/3) complex.
  • Arp2/3 actin-related protein 2/3
  • These ectopic actin filaments displace mitochondria from I- bands, cause aberrant fission and fusion balance, and impair respiratory chain activity.
  • neuronally- secreted vMSP promotes muscle mitochondrial localization and function, perhaps in an effort to modulate energy homeostasis.
  • vMSP signaling to muscle mitochondria might be relevant for the energy balance in ALS8 disease.
  • ALS8 patients studied five had increased cholesterol levels, four had reduced HDL, three had elevated triacylglycerol levels, and one was diabetic (Marques et al., 2006). More generally, ALS is associated with a spectrum of abnormalities in energy metabolism, including mitochondrial defects in neurons and skeletal muscle, insulin resistance,
  • the inventors show in C. elegans that loss of the VAP homolog VPR-1 causes triacylglycerol (TAG) accumulation in striated body wall muscle.
  • TAG triacylglycerol
  • Mosaic analysis and tissue-specific expression studies demonstrate that VPR-1 acts in neurons, not muscle to regulate muscle fat levels. Restoring muscle mitochondrial localization and function in vpr-1 mutants decreases muscle fat content. The fat metabolism alterations are part of a compensatory response mediated by the DAF-16/FoxO transcription factor. FoxO promotes muscle fat accumulation, maintains ATP levels during aging, and extends lifespan without influencing muscle mitochondrial morphology, localization, or function.
  • the inventors provide evidence that skeletal muscle metabolism is abnormal in Vapb mutant mice. The results indicate in the model that disrupting vMSP signaling triggers a compensatory response to muscle mitochondrial dysfunction involving FoxO transcripton factors.
  • vpr-l/vap loss increases fat levels in adult body wall muscle and intestine
  • vpr-1 mutant hermaphrodites In studies of vpr-1 mutant hermaphrodites, the inventors considered that body wall muscles often contain large lipid-like droplets not observed in wild-type controls (FIG. 8A). These lipid-like droplets were visible by differential interference contrast (DIC) and transmission electron microscopy (TEM). TEM of vpr-1 (tml411) null mutant muscle shows an expanded muscle belly filled with mitochondria, as previously reported (Han et ah, 2012), and large droplets (FIG. 8B). Large droplets were not observed in TEM of wild- type muscle, vpr-1 mutants also have darker intestines than those of wildtype hermaphrodites. Dark colored intestines are seen in mutant worms with high TAG content (Watts, 2009).
  • DIC differential interference contrast
  • TEM transmission electron microscopy
  • fluorescent compounds can be used to directly visualize fat stores in live tissue (Kubagawa et al., 2006; Klapper et al., 2011).
  • wild-type hermaphrodite controls dietary Bodipy-FAs were observed primarily in the intestine with a few small droplets present in muscle.
  • muscles of vpr-1 (tml411) null mutants contained numerous large Bodipy-FA-stained droplets (FIG. 8C). These fluorescent droplets perfectly overlapped with those observed in muscle by DIC microscopy (FIG. 8D).
  • Bodipy-FAs are continuously transported from the diet, to the worm' s intestinal cells, and then to the muscle, where they are tightly packed in membrane -bound vesicles.
  • Lipids were analyzed by electrospray ionization tandem mass spectrometry spectrometry (ESTMS/MS).
  • ESI-MS/MS analysis of the extracts detected a robust increase in TAGs in vpr-1 mutant extracts, but not in the membrane phospholipids phosphatidylethanolamine and phosphatidylcholine (FIG. 8E). These data indicate that loss of vpr-1 causes TAG accumulation in muscle and intestine of adult hermaphrodite worms.
  • VAP homologs have been implicated in ER stress pathways (Tsuda et al., 2008; Moumen et al., 2011; Gkogkas et al., 2008), which can modulate lipid metabolism and homeostasis (Basserti and Austin, 2012). Furthermore, mitochondrial dysfunction is sometimes associated with ER stress. The inventors considered the possibility that increased ER stress might cause the high muscle fat levels in vpr-1 mutants. Three lines of evidence argue against this possibility. First, an integrated hsp-4/BiPp::gfp ER stress reporter (Urano et al., 2002) did not show elevated stress levels in vpr-1 mutants (FIG. 18A).
  • vpr-1 mutants are not more sensitive than wild type to tunicamycin treatment, which induces ER stress (FIG. 18B).
  • vpr-1 is ubiquitously expressed and its homologs have been implicated in regulating lipid dynamics via a cell autonomous mechanism (Lev et al., 2008; Wyles et al., 2002; Jansen et al., 2011; Peretti et al., 2008).
  • VPR-1 functions to regulate muscle fat
  • the inventors first used genetic mosaic analysis.
  • Transgenic vpr-1 (tml411 ) mutant hermaphrodites were generated containing the vpr-1 genomic locus and the lineage marker sur-5::GFP expressed from an extrachromosomal array (Yochem and Herman, 2003). In C.
  • mosaic worms can be generated with losses in neurons, body wall muscles, intestinal cells, and the germ line.
  • vpr-1 genomic locus in vpr-1 (tml411 ) null worms rescued the fat metabolism defect in muscle (FIG. 9), as well as the muscle mitochondrial defects, sterility, slow growth, and other phenotypes.
  • Body wall muscles are generated from multiple cell lineages, including the EMS lineage. Transgene array loss in the EMS lineage generates mosaic worms that have a subset of muscles lacking vpr-1 expression. These muscle cells exhibited low fat levels, identical to muscle cells that express vpr-1 (FIG. 9). Therefore, VPR-1 is not required in body wall muscle.
  • VPR- 1 acts cell nonautonomously in neurons and germ cells (or their differentiation products) to modulate fat levels in muscle.
  • vpr-1 null mutants are sterile, due to a failure of germ cells to differentiate into sperm and oocytes.
  • MSPs that may influence fat metabolism
  • VAPB P56S mutation acts as a dominant negative by inhibiting secretion of the wild-type and mutant vMSPs (Tsuda et ah, 2008; Han et ah, 2012).
  • the inventors generated transgenic worms expressing P56S VPR-1 under the unc-119 neuronal promoter.
  • DAF-16/FoxO is required for fat accumulation in vpr-l/vap mutants
  • DAF-16/FoxO transcription factor which controls expression of genes involved in fat synthesis, fat transport, ⁇ -oxidation, and stress resistance
  • DAF-16 affects fat metabolism in vpr-1 mutants they generated vpr-l(tml411) daf-16(mu86) double mutants. Muscles of daf-16(mu86) null mutants contain few Bodipy-FA-stained droplets, similar to muscles of wild-type controls.
  • muscle fat levels in the double mutants were also low, and strongly reduced when compared to those in vpr- l(tml411 ) mutants alone (FIG. 12A).
  • daf-16 loss did not affect food intake, assessed by measuring pharyngeal pumping rates (FIG. 12B; P > 0.05), muscle mitochondria (see below), or sterility of vpr-l(tml411) mutants.
  • the elevated fat levels in vpr-1 null mutants require DAF-16/FoxO activity.
  • sod-3p::GFP sod-3 promoter
  • a direct DAF-16 target Oh et al., 2006; Henderson et al., 2006.
  • VPR-1 may regulate DAF-16 independent of insulin signaling.
  • DAF-16::GFP translocation to the nucleus is observed in stressed mutant and wild-type worms, suggesting that the insulin pathway is functional (FIGS. 13A and 13B).
  • vpr-1 mutants appear more sensitive to higher temperatures that require increased metabolic activity.
  • DAF-16/FoxO increases ATP levels and extends lifespan of vpr-l/vap mutants
  • ATP levels in daf-16 mutants are similar to wild-type controls, as previously shown (Houthoofd et ah, 2005; Braeckman et ah, 2002). These data indicate that DAF-16/FoxO helps vpr-1 mutants maintain ATP levels during aging.
  • vpr-1 mutants Based on the abnormalities in energy metabolism, the inventors tested whether DAF-16 influences lifespan in vpr-1 mutants. Similar to other worm mutants with mild or tissue-specific reduction in mitochondrial function, vpr-l(tml411 ) mutants have slightly extended adult lifespan compared to wild-type worms (FIG. 15B; mean adult lifespan + S.D. of
  • Vapb knockout mice exhibit signs of abnormal skeletal muscle energy metabolism
  • Vapb -/- mice do not exhibit overt defects in energy metabolism. In particular, body weight and glycemia appear normal with age. However, an energy metabolism defect of Vapb deficient mice might be unmasked by modifying insulin supply through feeding and fasting paradigms. In worms and mice, fasting reduces insulin signaling and increases FoxO activity, resulting in altered metabolic gene expression. The inventors used Vapb -/- mice of 2-6 months of age to avoid any confounding effect of the motor dysfunction observed at 18 months(Kabashi et ah, 2013).
  • mice were either fasted for 24 hours (fasted group) or fasted for 16 hours and re-fed for 8 hours to synchronize meals (fed group).
  • fasting decreased the TAG levels in the gastrocnemius (GA) muscle (FIG. 16A; P ⁇ 0.05).
  • TAG levels remained unchanged upon fasting in Vapb -/- GA and tibialis anterior muscles (FIG. 16A).
  • TAG levels were unchanged upon fasting and feeding in either +/+ or -/- mice (FIG. 16A).
  • Vapb ablation increases the resistance of muscle lipid stores to fasting induced mobilization.
  • the inventors next looked at mRNA levels of metabolic genes by quantitative RT-PCR.
  • Vapb ablation potentiated induction of the direct FoxOl target gene phosphoenolpyruvate carboxykinase (PEPCK) in response to fasting, but had no effect on fasting induction of other FoxOl targets such as glucose 6-phosphatase (G6Pase) and pyruvate
  • FIG. 16B 40530616.1 dehydrogenase kinase (PDK4) (FIG. 16B). FoxOl and Fox03 mRNA and proteins were similar in +/+ and -/- livers, and FoxOl up-regulation by fasting appeared normal in -/- liver (FIG. 16B).
  • VAPB mutant worms and mice appear to have muscle energy metabolism alterations, at least in part involving FoxO targets.
  • Results from Drosophila and C. elegans support the model that VAP MSP domains are secreted neurogenic factors that promote muscle oxidative metabolism (Han et ah, 2012).
  • C. elegans neurons cleave the vMSP and secrete it into the surrounding environment.
  • Secreted vMSPs signal via SAX-3 Roundabout and CLR-1 Lar-like receptors expressed in muscle, down-regulating Lar signaling to the Arp2/3 complex. This signaling pathway restricts actin filament formation to I-bands of the myofilaments, thereby localizing mitochondria to I- bands and promoting mitochondrial function (Tsuda et ah, 2008; Han et ah, 2012).
  • impaired vMSP signaling to muscle mitochondria triggers altered DAF- 16/FoxO transcription factor activity.
  • FoxO increases TAG synthesis and transport to muscle, helps maintain ATP levels during aging, and extends lifespan.
  • reduced vMSP signaling puts animals in an energy deficit, which triggers an altered metabolic response involving FoxO.
  • Evidence for this model and implications for ALS are discussed below.
  • vpr-l/vap loss triggers elevated DAF-16/FoxO activity in the presence of food (and insulin), resulting in muscle fat accumulation.
  • VPR-1 the regulation of FoxO by VPR-1 appears to be independent of insulin secretion.
  • FoxO transcription factors are critical regulators of energy metabolism, particularly under fasting conditions.
  • the inventors show that Vapb ablation in mice renders muscle lipid stores resistant to fasting, a situation analogous to lipid accumulation in vpr-1 mutant worm muscles. Indeed, muscle fat persists for long time periods in starved vpr-1 mutant worms.
  • Dysregulated lipid stores in mutant mice is associated with alterations in muscle gene expression consistent with abnormal FoxOl and Fox03 activity (Cheng and White, 2011).
  • FoxOl target gene mRNAs for PEPCK and G6Pase are clearly up-regulated in muscle of young Vapb -/- mice in the fed state ⁇ i.e. in the presence of insulin that decreases FoxOl activity).
  • a key finding in worms is that DAF-16/FoxO activity prolongs the adult lifespan of vpr-1 mutants from 6.9+2.5 to 12.9+4.4 days. This lifespan increase may be due to metabolic alterations that compensate for mitochondrial dysfunction. Consistent with this idea, FoxO extends the lifespan of C. elegans with reduced mitochondrial function (Dillin et al., 2002; Lee et al., 2003; Rea et al., 2007). The FoxO-dependent fat accumulation in vpr-1 mutant muscle may reflect an effort to increase energy production. The inventors show that DAF-16 helps vpr-1 mutants maintain ATP levels in 3-day old adults.
  • DAF-16 metabolic genes are those involved in fat synthesis and transport, ⁇ -oxidation, the glyoxylate cycle, and gluconeogenesis (Murphy, 2006).
  • additional DAF-16 targets may also be involved, such as stress resistance enzymes (Murphy, 2006; Murphy et al., 2003; Nissan and Honda, 1999).
  • vpr-1 mutants are more resistant than the wild type to reactive oxygen species and ER stress. Based on identified DAF-16 targets and vpr-1 mutant phenotypes, DAF-16 might increase energy substrate availability in muscle, stimulate anaerobic metabolism, increase oxidative metabolism in non-muscle cells, or decrease ATP consumption. Further studies are necessary to distinguish among these possibilities, as well as other models.
  • Vapb loss in mice does not cause motor neuron degeneration (Kabashi et ah, 2013; Han et ah, 2012), providing strong evidence that mitochondrial and metabolic defects are not secondary consequences of neurodegeneration. These data contrast with a recent Drosophila study suggesting that VAPB loss causes neurodegeneration via increased phosphoinositides (Forrest et ah, 2013). In humans, metabolic alterations caused by VAPB loss may not be sufficient to induce motor neuron degeneration, although they could strongly predispose to ALS. Redundancy could be an important consideration in the different models.
  • the worm genome encodes a single vap homolog, but many genes with MSP domains. Vertebrate genomes typically encode VAPA and VAPB, which are approximately 60% identical. Vap mutant flies have the most severe developmental defects and the fewest MSP genes in the genome.
  • striated muscle mitochondrial dysfunction alters FoxO activity, which in turn affects energy metabolism and promotes survival.
  • reduced vMSP signaling causes some of the mitochondrial and metabolic alterations in ALS patients.
  • vMSPs protect against ALS via effects on skeletal muscle energy metabolism.
  • C. elegans Bristol N2 is the wild-type strain. Worms were grown on NGM plates with NA22 bacteria as the food source (Brenner, 1974). Strain construction and marker scoring were done as previously described (Han et ah, 2012; Miller et ah, 2013).
  • strains and genetic markers used or generated were as follows: CF1553 muIs84[pAD76(5 , o ⁇ i-J.- :GFP)], CF1038 daf-16(mu86) I, vpr- l(tml411 )/ hT2[bli-4(e937) let-?(g782) qls48] LTII, SJ4005 zcls4[hsp-4::GFP], TJ356 zls356[daf-16p::daf-16::GFP; rol-6] IV, and XM1004 vpr-l(tml411) daf-16(mu86)l KT2[bli-4(e937) ⁇ et-l(q782) qls48] I;III.
  • RNAi was performed using the feeding method starting at the LI stage, as previously described (Han et ah, 2012).
  • arx-2 and xbp-1 RNAi clones are from the genome- wide library (Kamath and Ahringer, 2003). Each clone was sequenced for confirmation.
  • the marker plasmids pRF4 [rol-6] (60 ng/ ⁇ ) or myo-3p::mito::GFP (60 ng/ ⁇ ) were mixed with myo-3p::vpr-l (60 ng/ ⁇ ), ges-lp::vpr- 1(60 ng/ ⁇ ), unc-119p::vpr-l (60 ng/ ⁇ ), or unc-119p::vpr-l P56S (60 ng/ ⁇ ) and microinjected into the gonads of young adult hermaphrodites. Injected worms were incubated for 24 hours, transferred to new NGM plates, and screened for transgenic progeny.
  • Transgenic lines were selected based on the roller phenotype or GFP expression. Multiple independent transgenic lines were generated for all strains. To conduct genetic mosaic analysis, 10 ng/ ⁇ WRM06B28 fosmid DNA containing the vpr-1 genomic locus was mixed with 10 ng/ ⁇ pTG96 (sur-5p::GFP) plasmid and microinjected into the gonads of vpr-l(tml411 )ftiT2 hermaphrodites. Transgenic lines were selected based on GFP expression. Transgenic lines were maintained as vpr-l(tml411 ) homozygotes, as the fosmid rescued the sterility, mitochondria, fat metabolism, slow growth, and embryonic defects.
  • Transgene loss in the AB lineage was scored by GFP loss in head and tail neurons, the nerve cords, and the excretory gland.
  • Transgene loss in the PI lineage was scored by GFP loss in the intestine, muscle, somatic gonad, and hypl 1.
  • the P2 lineage was scored by GFP loss in numerous body wall muscle cells and hypl 1
  • the P3 lineage was scored by GFP loss in body wall muscle
  • the P4 lineage was inferred by a sterile phenotype without GFP loss.
  • Transgene loss in the EMS lineage was scored by GFP loss in the intestine and somatic gonad, while loss in the E lineage was score by exclusive GFP loss in the intestine.
  • Bodipy-FA Molecular probe, U.S. A
  • worms were washed five times with 70% ethanol.
  • wild type and vpr-l(tml411 ) mutants were processed in the same tube and identified based on gonad morphology.
  • lipids from equal masses of wild type and vpr-l(tml411 ) mutant adults were extracted by chloroform-methanol following a modified Bligh/Dyer extraction (Bligh and Dyer, 1959).
  • a mixture of internal standards including T17: l TAG was added to the chloroform-methanol phase before extraction.
  • the extracted samples were concentrated to dryness under a nitrogen stream, reconstituted with methanokchloroform (1: 1 v/v) and transferred to HPLC auto samplers.
  • Lipids were analyzed by ESI-MS/MS using an API 4000 (Applied Biosystems/MDS Sciex, Concord, Ontario, Canada) triple quadrupole mass spectrometer.
  • Extracted lipid samples (5 ml) were infused into the mass spectrometer with a solvent mixture of chloroform-methanol (1:2, v/v) containing 0.1% formic acid using a Shimadzu Prominence HPLC with a refrigerated auto sampler (Shimadzu Scientific Instruments, Inc.
  • worms were stained using the MitoTracker CMXRos dye (Molecular Probes, U.S.A), as previously described (Han et ah, 2012).
  • This lipophilic cationic fluorescent dye accumulates in mitochondria in a membrane potentialdependent manner (Gilmore and Wilson, 1999).
  • L4 larval stage worms were placed on dried plates containing a 100 ⁇ MitoTracker CMXRos dye solution (dropped on bacteria). After 24 hours incubation in the dark, worms were transferred to a new NGM plate and incubated in the dark for 20 minutes to remove intestinal background. Worms were mounted on dried 2% agarose pads without anesthetic. Wild-type and vpr-l(tml411 ) mutant hermaphrodites were cultured on the same plates.
  • ATP concentration was measured as described previously, with slight modification (Han et ah, 2012). Briefly, 150 worms were individually picked and placed into tubes containing M9 buffer, washed four times, and incubated at 20°C for 40 minutes to remove intestinal bacteria. These worms were then washed four times with TE solution (100 mm Tris-Cl, pH 7.6, 4 mm EDTA) and placed into microfuge tubes containing 300 ⁇ TE solution. Worm extracts were prepared by a series of cycles including freezing, thawing, and sonicating. These extracts were boiled for 10 minutes to release ATP and block ATPase activity.
  • Carcasses and insoluble material were pelleted in a microcentrifuge at 20,000 x g for 10 minutes.
  • the soluble extracts were diluted in a 1: 10 ratio using TE solution.
  • ATP concentration in 60 ⁇ of diluted extracts was measured using the ENLITEN ATP Assay System (Promega, U.S.A), according to the manufacturer's instructions.
  • a luminometer (Berthold, Germany) was used for quantification. Protein concentration was determined using the BCA protein assay (Pierce, U.S.A). ATP measurements were repeated at least three times for each strain.
  • Oxygen consumption rates were measured as previously described using the oxygraph system (Hansatech, UK) with minor modifications (Han et ah, 2012). Worms were cultured at 20°C and synchronized to the 1 -day-old adult stage. For each test, 1000 worms were individually picked and placed into a glass tube with 1 ml M9 buffer at 20°C. Collected worms were incubated for 40 min at 20°C to remove intestinal bacteria, carefully washed five times, and placed into 1ml M9 buffer. The worm solution was loaded into the chamber equipped with a SI Clark type polarographic oxygen electrode disc maintained at 20°C. Oxygen concentration was measured for 10 minutes. For normalization, worms were carefully collected from the chamber and protein content was measured using the BCA test kit (Pierce, U.S.A.). Rates were normalized to either total protein content or number of worms. At least three independent measurements were performed per strain.
  • worms were cultured at 20°C and 1 -day-old adult worms were placed on new NGM plates. Feeding behavior was recorded using a Zeiss Lumar stereomicro scope with AxioCam MRM digital camera. Measurements were conducted during a 30 second period at room temperature (22°C). The rhythmic contractions of the pharyngeal bulb were counted. For each strain, over 20 worms were counted. To determine lifespan of worms, L4 larval stage worms were placed on new NGM plates seeded with NA22 bacteria and cultured at 20°C.
  • the L4 stage was used because a small percentage of vpr-1 mutants die during L1-L4 stages and vpr-1 mutants develop slowly. Worms were monitored every day and transferred to flesh NGM plates. Death was scored by failure to respond to touching with a platinum wire. Wild-type worms fed NA22 bacteria have slightly shorter lifespan than worms fed OP50 bacteria.
  • mice were used and genotyped as described(Kabashi et ah, 2013). Mice (8-10 per group) were either fasted for 24 hours from 5PM (fasted group), or fasted from 5PM to 9AM and refed until sacrifice at 5PM. Liver and muscle GA and tibialis anterior (TA) tissues were collected, and
  • liver and TA muscle were incubated in Lysis buffer containing complete protease and phosphatase inhibitor cocktails. Protein concentration was measured using BCA Protein Assay. Equal amount of protein (50 ⁇ g) were separated by SDS- PAGE 10% and blotted onto nitrocellulose membrane. Membranes were saturated with 10% milk and then incubated with the primary antibodies FoxOl (ProteintechTM; 18592-1-AP), Fox03a (Cell signaling; #2497), VAPB [4] and Histone H3 (Cell signaling; #9715), all diluted (1: 1000) followed by anti-rabbit secondary antibody, diluted 1:5000.
  • FoxOl ProteintechTM; 18592-1-AP
  • Fox03a CD34
  • VAPB [4] Histone H3
  • tissue powder was homogenized in lysis buffer (250 mM Sucrose solution, 1 mM EDTA, 2% SDT, 1 mM DTT, 10 mM Tris HC1 pH 7.4) containing protease inhibitors (Sigma P8340) and phosphatase inhibitors (Sigma 8345), centrifuged at 12000 x rpm for 15 minutes at room temperature.
  • TAG concentration was determined in duplicate for each sample in 5 ⁇ of supernatant, using the enzymatic method of analysis (Randox Triglyceride Colorimetric Assay Kit, Randox Laboratories Limited, UK) as described by the manufacturer. Lipid values were normalized to protein concentration.
  • a familial form of Amyotrophic lateral sclerosis is caused by a point mutation (P56S) in the VAMP associated protein B (VapB).
  • VapB VAMP associated protein B
  • Human VapB and Drosophila Vap- 33-1 Vap are homologous type II transmembrane proteins that are localized to the ER.
  • Vap is required for ER protein quality control (ERQC).
  • Loss of Vap in flies shows various ERQC associated defects, including protein accumulation, ER expansion, and ER stress. It is also shown
  • Oxysterol binding protein (Osbp). Although loss of Osbp does not show obvious phenotypes, except male sterility, restoring expression of an Osbp that does not require Vap binding suppresses the ER defects caused by loss of Vap. Hence, in embodiments of the disclosure, the ALS8 mutation impairs the interaction of Vap with Osbp, resulting in hypomorphic defects that are similar but less severe than those observed in the vap null mutant.
  • ALS Amyotrophic lateral sclerosis
  • ALS8 is an autosomal dominant form of ALS caused by a point mutation (P56S) in the gene encoding the VapB protein (Nishimura et ah, 2004). Human vapB is evolutionarily conserved, with homologs in numerous species (Lev et ah, 2008), including Drosophila that contains Vap-33-1 or vap.
  • Vaps contain an amino (N)-terminal domain, called the major sperm protein (MSP) domain (Nishimura et ah, 1999; Weir et ah, 1998) and a transmembrane domain that anchors the protein in the ER (Kaiser et ah, 2005; Skehel et ah, 2000; Soussan et ah, 1999).
  • MSP major sperm protein
  • Vaps most likely also have autonomous functions as they are ER associated proteins. Indeed, they have been shown to function in glucose transport (Foster et ah, 2000), neurite extension (Matsuzaki et ah, 2011) and the development of the neuromuscular junctions (Pennetta et ah, 2002). Importantly, Vaps have also been implicated in the regulation of phospholipid biosynthetic proteins (Peretti et ah, 2008). Vaps interact with proteins containing two
  • phenylalanines in an acidic tract FFAT
  • Motova and Levine, 2012 phenylalanines in an acidic tract
  • Oxysterol binding protein Osbp
  • Cet ceramide transport protein
  • Studies with cultured cells indicate that the Vap/Osbp interaction is required for sphingomyelin (SM) biosynthesis in response to 25- hydroxycholesterol (Lagace et ah, 1999; Peretti et ah, 2008; Perry and Ridgway, 2006).
  • SM sphingomyelin
  • the ER is the site where newly synthesized proteins are folded and modified. Protein folding in the ER is monitored by a stringent ER quality control (ERQC) system that only permits properly folded proteins to traffic to the Golgi (Araki and Nagata, 2011; Balch et ah, 2008; Braakman and Bulleid, 2011).
  • ERQC ER quality control
  • the accumulation of misfolded proteins in the ER caused by alterations in ER homeostasis initiates ER stress that attempts to resolve the protein-folding defects (Friedlander et ah, 2000; Travers et ah, 2000).
  • ER stress has been observed in human sporadic ALS patients (Atkin et ah, 2008) and in SOD1 transgenic mice (Nishitoh et ah, 2008; Saxena et ah, 2009).
  • Overexpression of the ALS8 mutant Vap causes ER stress in flies (Tsuda et ah, 2008).
  • studies with cultured cells showed that overexpression of the ALS8 mutant Vap inhibits ER stress (Gkogkas et ah, 2008; Kanekura et ah, 2006).
  • the precise role of Vap in ER biology remains to be determined.
  • Vap is required for ER protein homeostasis. Loss of Vap causes defects in ERQC, resulting in protein accumulation and ER stress. Loss of Vap also causes Osbp to be mislocalized from ER to Golgi, and restoring expression of Osbp in the ER partially suppresses the defects caused by loss of Vap. In certain embodiments of the disclosure, loss of Vap contributes to ER stress and that this stress might play a role in the pathology of ALS.
  • the genomic ALS8 mutant vap construct was created by chimeric PCR with primers containing a P58S mutation. The genomic fragment was subcloned into the pCasper 4 vector. To create site specific transgenes (Venken et al., 2006), the attB sequence was inserted in the genomic wild type (WT) or ALS8 mutant vap/pCasper 4 constructs. The attB constructs were injected into VK31 and VK33 attP docking sites (Venken et al., 2006).
  • vap A31 was created by imprecise excision of the P-element present in y w, P ⁇ Mae-UAS.6.11J, Vap-33-lGG01069 (Bellen et al., 2004).. Southern blotting and western blotting with the Vap (Rb92) antibody (Tsuda et al., 2008) shows that [vap A31] is a null allele.
  • the stock containing the precise excision of y w, P ⁇ Mae-UAS.6.11J,Vap-33-lGG01069 was used as a control.
  • the osbp null mutant was created by screening for a deletion of the DNA between two piggyBac elements, PBac ⁇ WHJOsbpf00496 and PBac(RBJe04437 (Thibault et al, 2004) using the method described by Golic and Golic (Golic and Golic, 1996) .
  • a domain of the Osbp protein (aa 60-400) was expressed using the GST- fusion protein system.
  • a polyclonal Guinea Pig antibody (GP89) was raised against the fusion protein at Cocalico Biologicals (Reamstown, PA). This antiserum was used at 1:3000.
  • Larvae and adult brains were fixed in 4% paraformaldehyde for 20 minutes and washed with PBS containing 0.2% Triton X-100. The following antibody dilutions were used: rabbit anti- Vap (RB92)(Tsuda et al., 2008), 1: 1000; mouse monoclonal anti- Chaoptin (Van Vactor et al., 1988), 1:200; Elav
  • guinea pig anti-Bip (Ryoo et al, 2007), 1:500 (Ryoo et al, 2007) and mouse anti-Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol ⁇ Kidd, 1998 #107)(Ryoo et al, 2007) and mouse anti- Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol ⁇ Kidd, 1998 #107)and mouse anti-Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol (Kidd et al, 1998), 1:200 and anti-N-Cadherin (Iwai et al, 1997), 1:200. Secondary antibodies conjugated to Cy3 or Alexa 488 (Jackson ImmunoResearch, Molecular Probes) were used at 1: 1000. Images were captured with a Zeiss LSM510 confocal microscope and processed with ImageJ.
  • WT and ALS8 human Vap protein were subcloned into the pBD vector (Rual et al, 2005).
  • Human OsbpL8 and Orp3 were subcloned into pAD vector (Rual et al, 2005). All interaction assays were performed by co-transfecting the two vectors encoding the hybrid proteins into the yeast strain Mav203 and plating on selective media lacking histidine.
  • PGAL assays were performed as described previously (Rual et al, 2005).
  • Giant Fiber recordings were performed with a protocol modified from (Tanouye and Wyman, 1980). Briefly, flies were anaesthetized on ice, transferred to a petri dish filled with soft dental wax, and the fly wings and legs were mounted in wax, ventral side down, using forceps.
  • TTM TegoTrochanter Muscle
  • DLM Dorsolateral Muscle
  • five electrolytically sharpened tungsten electrodes were used: two for stimulating the giant fiber (GF), one as a reference electrode, and two for recording from the TTM and DLM respectively. To activate the GF, two sharp tungsten electrodes were inserted into each eye and voltage stimulation was applied at different frequency stimulations.
  • GF-DLM and GF-TTM responses were measured through two electrodes implanted in the DLM and TTM.
  • low frequency stimulations at 0.5Hz were applied after placing the two recording electrodes in TTM and DLM to ensure that the electrodes are recording from the proper muscles (the latency of responses for TTM is 0.8ms and for DLM 1.2ms; Tanouye and Wyman, 1980).
  • High frequency train stimulations of 20 pulses were delivered to the GF at 10Hz, 20Hz, 50Hz, 100Hz and 200Hz in random order. Ten times repetitive stimulations were applied for each particular frequency train, interspersed with five minutes rest between two trains of stimuli.
  • 0.5Hz stimulations were used again after high frequency stimulation to confirm that electrodes were still in the proper muscle.
  • Stimuli of the crossing electrodes were fixed at a duration of 0.1ms at 8-15 V of amplitudes through a stimulus isolation unit (model DS2A, Digitimer Ltd, England) and the frequency of train stimuli was controlled by AxoGraph acquisition software (AxoGraph Scientific).
  • a microelectrode amplifier (Model 1800, A-M system) was used for all recordings.
  • Digidata 1322A Anaxon Instruments was used for data acquisition. The probability of responses, under particular frequency of GF
  • the difference of probability of responses between control and mutant (p value) for particular stimuli were calculated by a t-test (SigmaPlot 10; Systat Software, Inc.).
  • MSP Vap The MSP domain of Vap (MSP Vap) is cleaved and functions as a secreted ligand for muscle expressed Eph, Robo, and Dlar receptors in flies and worms (Han et ah, 2012; Tsuda et ah, 2008). Vap is also localized at the ER and overexpression of the ALS8 mutant isoform causes ER stress in flies (Tsuda et ah, 2008), suggesting that Vap may play a role in ER stress as well. It was therefore examined if Vap is required for ER proteostasis.
  • the ER is integral to maintaining protein homeostasis (proteostasis), as protein-folding of transmembrane and secreted proteins occurs under the supervision of ERQC (Vembar and Brodsky, 2008).
  • the ERQC is able to identify misfolded proteins, retrotranslocate the misfolded proteins and promote their degradation.
  • ERQC overload induces ER stress, which restores proteostasis by halting protein translation, and by activating signaling pathways that lead to an increased production of molecular chaperones, which facilitate protein folding (Bernales et ah, 2006).
  • the ERQC is important especially for membrane proteins, which are prone to aggregation due to their inherent tendency to assemble in oligomers (Hurtley and Helenius, 1989).
  • CD8-GFP membrane anchored GFP
  • a broadly expressed neuronal GAL4 driver OK307-GAL4 driver
  • FIGGS. 2B, B' and B stained adult cortical neurons with anti-Chaoptin and anti-GFP antibodies
  • vacuoles are contiguous with the nuclear membrane (FIG. 22C, arrows) and decorated with electron-dense ribosomes (FIG. 22D, arrow heads), indicating that they correspond to a vastly expanded ER.
  • ER expansions are typically observed in the presence of ER stress (Schuck
  • Ubiquitinated proteins accumulate in subpopulation of cortical neurons of adult mutant neurons (FIG. 23H and H'), but not in brains of larvae and control adults (FIG. 4G and G'). Hence, the data suggest that loss of Vap leads to accumulation of ubiquitinated proteins.
  • VAP is required for the proper localization of OSBP
  • Neurons in which the ALS8 protein is overexpressed also exhibit an ER stress, similar to what is observed in the vap null mutant ((Tsuda et al, 2008) and FIG. 23).
  • the inventors therefore examined if the ALS8 mutation causes a loss of interactions with proteins that are required for its function.
  • the inventors performed a two hybrid screen using a human adult brain cDNA library to identify proteins that are able to bind to the WT human protein, but are unable to interact with the ALS 8 human Vap mutant protein when used as a bait (Rual et al, 2005).
  • OSBP related proteins ORP3 (Gregorio-King et al, 2001; Lehto et al, 2005; Lehto et al, 2008) and OSBPL6 (Lehto and Olkkonen, 2003). Both interact with wild type VapB, but are not able to interact with ALS 8 VapB as reported previously
  • the Drosophila Osbp homolog encoded by CG6708 contains a Vap binding site or FFAT-motif (Alphey et al, 1998; Ma et al, 2010). As shown in FIG. 24A,
  • Drosophila WT Vap can interact with Osbp in GST pull down assays, indicating that the interaction between Vap and Osbp is direct and evolutionarily conserved.
  • the Osbp protein family is an evolutionarily conserved protein family whose function was initially linked to non-vesicular intracellular transport of sterols in yeast (Raychaudhuri and Prinz, 2010; Ridgway, 2010; Yan and Olkkonen, 2008).
  • Osbps integrate sterol and sphingomyelin metabolism (Banerji et al, 2010; Goto et al, 2012; Yan and Olkkonen, 2008), as well as control of microtubule-dependent motility of
  • vap null mutant clones To determine whether Vap is required for the proper localization and function of Osbp the localization of Osbp was examined in vap null mutant clones.
  • the inventors performed MARCM analysis (Lee et al, 2000) to create mutant vap clones and labeled them with the Osbp antibody. As shown in FIG. 25, loss of vap (marked by GFP) leads to an aberrant distribution of Osbp and an accumulation of Osbp in punctae (arrows in FIG. 6B'). Co-staining the vap null mutant neurons with a Golgi marker, the Msl20 kDa antibody (Stanley et al, 1997)
  • ALS8 mutation causes a partial loss of function of Vap
  • VapALS8 ALS8 mutant Vap (VapALS8 ) in flies affects the secretion of the MSP aminoterminal domain of VapALS8, causing a loss of function of Vap. Furthermore, expression of VapALS8 causes an ER stress when overexpressed in neurons.
  • VAPALS8 is a partial loss of function mutation
  • transgenic flies carrying a genomic fragment of vap using P-element mediated transformation with and without a site specific attB docking site to attenuate positional effects of transgenes (Venken et ah, 2011).
  • the inventors first tested if the genomic vapWT can rescue the lethality associated with the vap null mutants (Avap) (FIG. 26B). Loss of vap causes pupal or pharate adult lethality with occasional adult escapers. All tested genomic vapWT transgenes (4 lines) rescue the lethality associated with loss of Vap and restore normal lifespan of the flies (FIGS. 26B and C). Eight out of eleven P-element vapALS8 transgenic lines also rescued the lethality, suggesting that VapALS8 protein retains some WT protein function (FIGS. 26B and C).
  • VapALS8 rescued flies exhibit a severely reduced lifespan when compared to VapWT rescued flies (FIG. 26B and C, compare Avap; vapWT (F7), 52 days and Avap; vapALS8 (M6), 23 days; * P ⁇ 0.001).
  • an additional copy of vapWT (Avap; vapWT ' / vapALS8 ) compensates for the defects associated with vapALS8 (FIG. 26B; compare the lifespan of Advap; vapALS8 (M6), 23 days and Avap ;vapWT/vapALS8 (M6), 65days; * P ⁇ 0.001), suggesting that the ALS8 mutation only causes a partial loss-of-function.
  • vapWT and vapALS8 transgenes were compared in the same docking site (VK31 line) to rescue the vap null associated phenotypes.
  • the inventors confirmed that vapALS8 rescued flies indeed have a severely reduced lifespan when compared to vapWT rescued flies (FIG. 26B,
  • vapALS8 (VK31 line), 14 days vs. ⁇ ; vapWT (VK31 line), 67days), showing that position effects are not responsible for the observed differences in life span.
  • FIG. 26D To assess the physiological consequences of the incomplete rescue flight ability (FIG. 26D), brain pathology (FIG. 27A), and the electrophysiological properties of adult neurons was examined using the giant fiber responses of Avap;vapALS8 (VK31) transgenic flies (FIGS. 27B and C). Interestingly, vapALS8 rescued flies exhibit a progressive flight defect (FIG. 26D) that worsens with age.
  • vapALS8 causes defects in flight ability in an age dependent manner.
  • FIG. 27A To assess the morphological consequences of the ⁇ flies rescued with vapALS8, histological sections were examined of adult brains that are 12 days old. As shown in FIG. 27A, there were very significant histological differences in these brains when compared to those of the proper controls. The adult brains of ⁇ ; vapWT flies did not show any obvious defects (FIG. 27A). In contrast, ⁇ ; vapALS8 flies exhibit numerous vacuoles in the optic lobe and central brain (FIG. 27A), but they are more frequent in the central lobe. Importantly, an additional copy of vapWT completely suppresses the defects associated with vapALS8, suggesting that the ALS8 mutation is less potent than the WT copy.
  • GFS giant fiber system
  • vap null mutants are unable to follow increasing stimulation rates, suggesting that loss of VAP causes severe functional defects in the adult motor neuron system (FIG. 27B).
  • the neuronal network must be intact as the flies still respond to a 10 Hz stimulation rather efficiently at day 1 when vap is lost.
  • vapWT transgene can fully rescue the defects in ⁇ adult motor neurons (FIG. 27C).
  • vapALS8 rescues the defects only partially (day 6) and exhibits a defect that progressively worsens ( ⁇ ; vapALS8, 6 days vs. 12 days), suggesting that vapALS8 causes a progressive demise of the GFS.
  • the data suggest that VapALS8 is less potent than VapWT and hence, the vapALSS mutation is a partial loss of function allele of vap. osbp genetically interacts with the ALS8 mutation
  • an impairment of the normal function of Vap in the ER may contribute to the pathology of ALS8.
  • Vap is required for the proper localization of Osbp in the ER.
  • the interaction between Vap and Osbp function is apparently required for the implementation of the ERQC.
  • Failure of Vap-Osbp to function causes a defect in ER proteostasis, resulting in protein accumulations in the ER and ER stress.
  • UPS Ubiquitin Proteasome System
  • patients express both WT Vap and the ALS8 mutant Vap. As the ALS8 mutant
  • Vap is not able to bind to Osbp, this loss of binding seems to result in partial loss of function of Vap. This in turn may cause a gradual decrease in function of the ERQC. These defects may also lead to secondary defects that have been previously reported: namely the lack of the secretion of MSP (Tsuda et al, 2008; Han et al, 2012).
  • VapB levels decrease concomitantly with the disease's progression in the SOD1 mouse model (Teuling et al., 2007), and sporadic ALS patients have been reported to have decreased levels of the VapB protein (Anagnostou et al., 2010; Teuling et al., 2007), suggesting that impaired VAPB function may contribute to the pathogenesis of familial and sporadic forms of ALS. It is therefore possible that the molecular mechanism by which loss of Vap causes defects may be not restricted to ALS 8.
  • VapB functions in protein homeostasis in the ER.
  • the ERQC is involved in identifying aberrantly misfolded proteins, retrotranslocating the misfolded proteins and processing the degraded retrotranslocated proteins (Vembar and Brodsky, 2008). These processes seem to be tightly linked. Indeed, many proteins
  • Vap is unlikely to function in chaperone-dependent refolding, since a molecular chaperone, Bip, is upregulated in vap null mutants. Moreover, Bip overexpression fails to rescue the ER stress in the vap null mutants.
  • Osbp Mammalian oxysterol binding protein
  • Orp OSBP-related protein
  • Osbp/Orp This domain organization suggests that a primary function of Osbp/Orp is to transfer cholesterol or oxysterols between target membranes, and/or transduce sterol-dependent signals at these points of contact (Banerji et ah, 2010; Ngo and Ridgway, 2009; Suchanek et ah, 2007).
  • Differential localization of Osbp between organelles in response to exogenous and endogenous sterol ligands suggests that Osbp transfers cholesterol and/or oxysterols between these organelles.
  • ER membrane is cholesterol poor (3-6% of total lipids) (Lange, 1991), acute cholesterol depletion in culture medium impairs the mobility of membrane proteins and thus protein secretion from the ER in cultured cells (Ridsdale et ah, 2006).
  • the defects in ER proteostasis might be due to decreased levels of cholesterol in the ER caused by loss of Osbp/Vap function.
  • Osbps are also coupled to the activation of ceramide transport protein (Cert) and sphingomyelin (SM) synthesis through increased activity of PI4KIIa, a cholesterol sensitive PI 4-kinase (Banerji et ah, 2010; Waugh et ah, 2006).
  • PI4KIIa a cholesterol sensitive PI 4-kinase
  • the mislocalization of Osbp may also affect the plasma membrane (PM), phosphatidylinositol 4-phosphate (PI(4)P) levels (Stefan et ah, 2011), the PM sterol distribution (Georgiev et ah, 2011) and polarized endocytosis (Alfaro et ah, 2011).
  • PM plasma membrane
  • P(4)P phosphatidylinositol 4-phosphate
  • loss of ALS8 and the ALS8 mutation cause similar phenotypes and the ALS8 mutation in patients is a partial loss of function mutation, i.e. VapB is haploinsufficient in humans.
  • studying the normal function of the ALS8 gene has advanced the understanding of the molecular, physiological, and pathophysiological aspects of the disease, and ER stress plays a role in numerous forms of ALS.
  • a model shows that MSP is secreted in blood and binds to growth cone guidance receptors, including ROBO and a LAR like phosphor- tyrosine phosphatase, to modulate muscle mitochondrial dynamics. It has been shown that ALS patients have reduced level of VapB and that these correlate with neuronal and muscle impairments. Therefore, it was considered that MSP serves as a hormone to control the health or disease status of neurons and muscles, in particular embodiments.
  • human HEK293 cells were treated with purified recombinant His-tagged MSP (His-MSP), and the overall phosphor- tyro sine profile was examined through western blotting.
  • MSP treatment promotes growth of the cultured primarily hippocampal neurons. There was an up-regulation in the bassoon positive synapses in these primarily cultured neurons upon His-MSP treatment (FIG. 30), indicating that the biological activity of MSP regulates synapse formation and/or maintenance in neurons, in certain embodiments of the disclosure. Taken together, the data indicates that MSP modulates complex protein phosphorylation signaling networks that control synapse formation in hippocampus neurons.
  • HEK293 cells were washed twice with ice-cold 1XPBS and then lysed in Lysis Buffer (20mM Hepes pH7.5, 150mM NaCl, 1%NP40, 50mM NaF, ImM Na3V04, 10% glycerol, protease inhibitor cocktail from Roche).
  • Lysis Buffer (20mM Hepes pH7.5, 150mM NaCl, 1%NP40, 50mM NaF, ImM Na3V04, 10% glycerol, protease inhibitor cocktail from Roche).
  • Anti-phosphotyrosine antibody (clone 4G10; Millipore) was used as first antibody to detected phosphotyrosine proteins.
  • Protein Phosphorylation Microarray (PathScan RTK Signaling Antibody Array Kit; Cell Signal) is as follows:
  • Decant Array Blocking Buffer by gently flicking out the liquid into a sink or other appropriate waste receptacle. Add 150 ⁇ diluted lysate to each well and cover with sealing tape. Incubate for 2 hours at room temp (or overnight at 4°C) on an orbital shaker.
  • STRING9.05 program http://string-db.org/ was used. The proteins hits isolated from the microarray assays were input to the program. The default settings were used to generate the signaling network.
  • the present example describes studies utilizing a polynucleotide encoding VAPB for therapeutic application in a mouse model for ALS. Described herein are two main outcome measures that showed improvement following viral expression of VAPB in the brains of mice of mouse model of ALS.
  • FIG. 31 illustrates an exemplary viral expression vector for injection into the SOD1 G93A transgenic mouse, wherein the construct encodes full- length VAPB and a label, such as yellow fluorescent protein (YFP), for example.
  • YFP yellow fluorescent protein
  • MSP domain of VAPB or a functionally active fragment or derivative of MSP are injected into the SOD1 G93A transgenic mouse.
  • FIG. 32 illustrates an example of an injection regiment for studies in the ALS mouse model.
  • RotaRod testing was used to assess outcome. In RotaRod testing, mice are placed on a rod and the rod is rotated
  • Time may be recorded either when the mice fall or when they rotate twice around the rod.
  • more than one trial per day may be given to the mice, and multiple training days may be included in a given week.
  • three trials a day were given for experimental and control mice, including for three training days, followed by once a week thereafter. Five-minute trials were performed, with acceleration of the rod from 5-40 rpm, with no reverse motion.
  • FIG. 33 shows RotaRod testing for control and SOD mice given AAV8- VAPB or left untreated. The decline in motor performance measured by rotarod testing is temporarily slowed in the mice treated with VAPB.
  • FIG. 34 illustrates survival rate of
  • VAPB/SOD1 G93A transgenic mice wherein the experimental mice had an extended life compared to control mice.
  • a second measure included a behavioral score, which is a crude measure of limb control used in the art.
  • the scores utilized for measure (described by The Jackson
  • FIG. 35 shows exemplary behavioral scores in VAPB-treated SOD mice compared to untreated SOD control mice lacking VAPB overexpression.
  • the VAPB-treated SOD mice decline more slowly at early points of disease than untreated SOD controls.
  • FIG. 36 shows that there is strong overexpression of VAPB protein in the brain of SODl/VAPB transgenic mice, and FIG. 37 demonstrates increased levels of MSP fragment in the plasma of wild-type mice treated with the same AAV8-VAPB vector.
  • Kesl/Osh4p is a regulator of polarized exocytosis. Traffic 12, 1521-1536.
  • OSBP oxysterol binding protein
  • VAP Vesicle associated membrane protein B
  • Oxysterol binding protein-dependent activation of sphingomyelin synthesis in the golgi apparatus requires phosphatidylinositol 4-kinase Ilalpha. Mol Biol Cell 21, 4141-4150.
  • 40530616.1 project single transposon insertions associated with 40% of Drosophila genes. Genetics 167, 761-781.
  • IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415, 92-96.
  • VAPB interacts with and modulates the activity of ATF6. Human molecular genetics 17, 1517-1526.
  • ORP-3 a human oxysterol-binding protein gene differentially expressed in hematopoietic cells. Blood 98, 2279-2281.
  • VAPB/ALS8 amyotrophic lateral sclerosis-linked P56S mutation of vesicle-associated membrane protein- associated protein B
  • Kikuchi H., Aimer, G., Yamashita, S., Guegan, C, Nagai, M., Xu, Z., Sosunov, A.A., McKhann, G.M., 2nd, and Przedborski, S. (2006).
  • OSBP-related proteins a novel protein family involved in vesicle transport, cellular lipid metabolism, and cell signalling. Biochimica et biophysica acta 1631, 1-11.
  • OSBP-related protein 3 ORP3
  • VAP protein family from cellular functions to motor neuron disease. Trends in cell biology 18, 282-290.
  • Protrudin serves as an adaptor molecule that connects KIF5 and its cargoes in vesicular transport during process formation. Mol Biol Cell 22, 4602-4620.
  • Oxysterol binding protein-related Protein 9 is a cholesterol transfer protein that regulates Golgi structure and function. Mol Biol Cell 20, 1388-1399.
  • VAMP-associated membrane protein-associated (VAMP-associated) proteins Molecular cloning and characterization of mammalian homologues of vesicle-associated membrane protein-associated (VAMP-associated) proteins. Biochemical and biophysical research communications 254:21-26.
  • ALS-linked mutant SOD1 induces ER stress- and ASKl-dependent motor neuron death by targeting Derlin-1. Genes & development 22, 1451-1464.
  • Saccharomyces cerevisiae CNE1 encodes an endoplasmic reticulum (ER) membrane protein with sequence similarity to calnexin and calreticulin and functions as a constituent of the ER quality control apparatus.
  • ER endoplasmic reticulum
  • Oxysterol-binding protein and vesicle- associated membrane protein-associated protein are required for sterol-dependent activation of the ceramide transport protein. Mol Biol Cell 17:2604-2616.
  • ERG30 a VAP-33-related protein, functions in protein transport mediated by COPI vesicles. The Journal of cell biology 146, 301-311.
  • Regulators of mitotic arrest and ceramide metabolism are determinants of sensitivity to paclitaxel and other chemo therapeutic drugs.
  • VAP vesicle-associated membrane protein-associated protein
  • VAPB amyotrophic lateral sclerosis 8 protein
  • EPHA4 is a disease modifier of amyotrophic lateral sclerosis in animal models and in humans. Nat Med 18, 1418-22.
  • VAP-A Vesicle-associated membrane protein- associated protein-A

Abstract

Amyotrophic Lateral Sclerosis is a progressive motor neuron-degenerative disease that results in loss of voluntary muscle control. Impaired VapB function contributes to the pathology of ALS through reduced secretion of MSP, the 125 amino acid N-terminal domain of VapB. Administration of MSP or fragments or derivatives thereof rescues complications associated with reduced MSP secretion and is used to treat individuals with a neurological disorder or suspected of having a neurological disorder, such as ALS.

Description

MSP AND ITS RECEPTORS IN THE THERAPY OF AMYOTROPHIC LATERAL
SCLEROSIS
[0001] This application claims priority to U.S. Provisional Patent Application Serial No. 61/801,218, filed March 15, 2013, and to U.S. Provisional Patent Application Serial No. 61/847,889, filed July 18, 2013, both of which applications are incorporated herein by reference in their entirety.
TECHNICAL FIELD
[0002] Embodiments of the disclosure are directed at least to the fields of neurobiology, molecular biology, cell biology, neuropharmacology, and medicine.
BACKGROUND
[0003] Amyotrophic Lateral Sclerosis (ALS) is a neurological disease that affects motor neurons in the brain and spinal cord. ALS is characterized by the progressive degeneration and death of motor neurons that control voluntary muscle activity. Reduced synaptic transmission to muscles causes initial symptoms that include twitching, cramping, muscle weakness and stiffness, slurred speech and difficulty swallowing, which gradually worsen as the disease progresses. Late stage symptoms include decreased strength and muscular control, muscular atrophy, difficulty breathing, drooling, gagging, head drop, difficulty walking, and paralysis. Most patients diagnosed with ALS die within 5 years due to cachexia (wasting syndrome) or respiratory failure.
[0004] Although the etiology of ALS is unknown, a common model proposes motor neurons are made vulnerable through environmental factors or genetic predisposition, and are injured by the excitatory neurotransmitter glutamate. Most motor neurons affected by ALS have a buildup of aggregates; however, it is unknown whether these aggregates are involved in causing ALS or are a byproduct of the dying cell. Familial ALS (FALS) which is responsible for 5-10 % of ALS cases is associated with mutations in several genes, including C9orf72, SOD1, TARDBP, FUS, ANG, ALS 2, SETX, and VapB genes. The majority of ALS cases are sporadic, for which no genetic association have been identified.
40530616.1 [0005] Given the multitude of symptoms presented by ALS, disease management requires individualized therapy and continual adaptation of medications. Various symptomatic treatments are available. Spasticity may be treated with sedative-hypnotics such as diazepam or skeletal muscle relaxants such as dantrolene. Uncontrolled muscle contractions may be treated with anticonvulsives such as carbamazepine. Pain resulting from uncontrolled muscle contractions may be treated with nonsteroidal anti-inflammatory drugs. Excess saliva and drooling may be treated with anticholinergic drugs such as atropine or scopolamine. The only drug that is currently labeled for treatment of ALS by the FDA is riluzole. Riluzole inhibits glutamate release, inactivates voltage-dependent sodium channels and interferes with intracellular events that follow transmitter binding at excitatory amino acid receptors. Riluzole does not halt the disease process, but has been shown to delay time to death by a few months. There exists a need for improved ALS therapeutic agents that go beyond symptomatic treatment and target the underlying mechanisms of ALS.
BRIEF SUMMARY
[0006] Embodiments of the disclosure concern methods and compositions related to the treatment and/or prevention of one or more neurological disorders. In particular
embodiments, the neurological disorder may be of any kind, but in specific embodiments the neurological disorder is amyotrophic lateral sclerosis (ALS). In certain aspects, a therapeutically effective amount of one or more compounds are provided to the individual diagnosed as having a neurological disorder or suspected of having a neurological disorder or at risk for having a neurological disorder. In specific embodiments, the neurological disorder is associated with impaired function of vesicle associated membrane protein (VAMP)-associated proteins (Vaps). In some embodiments, the neurological disorder is associated with a decrease in and/or mislocalization of VapB. In some embodiments, the neurological disorder is associated with a decrease in the secretion of the amino-terminal domain of VapB, named the MSP domain (Major Sperm Protein). In certain cases, the neurological disorder is associated with loss of MSP. In particular embodiments, delivery of Vap MSP to an individual with a neurological disorder allows for amelioration of at least one symptom of the disorder. In specific embodiments, the neurological disorder is ALS that may be of any type, including sporadic or familial ALS. In
40530616.1 particular embodiments, the disclosure concerns therapy treats VAPB-related ALS or SOD1- related ALS.
[0007] Thus, in specific embodiments, there are compositions and methods relating to the use of Major Sperm Protein (MSP), the N-terminal 125-amino acid domain (SEQ ID NO. 2) of the VapB protein (SEQ ID NO. 1), for the treatment of a neurological disorder, such as amyotrophic lateral sclerosis. In some aspects, compositions for treating ALS comprise a therapeutically effective amount of MSP. In particular embodiments, a functionally active derivative of MSP is provided to the individual, such as one that has one or more alterations compared to SEQ ID NO:2; in specific embodiments, there are one, two, three, four, five, six, seven, eight, nine, or ten or more alterations in the MSP derivative compared to SEQ ID NO:2. In particular embodiments, the MSP derivative lacks part of its N-terminus and/or C-terminus. In some cases, one or more internal amino acids are lacking compared to SEQ ID NO:2. One can determine whether or not the MSP derivative is functionally active by a variety of means such as, for example, testing for binding to its cognate muscle receptors (EphA4, Dlar, and/or Robo or human equivalents thereof).
[0008] The MSP or MSP derivative may be provided to the individual by any suitable means, such as intravenously, orally, subcutaneously, and so forth, although in specific cases there is peripheral delivery.
[0009] In some embodiments, there is a method for treating a patient with ALS comprising providing a therapeutically effective amount of a pharmaceutical composition of MSP to the patient. In specific embodiments, the MSP is provided to the patient multiple times. In some cases, the ALS is familial or sporadic. In particular embodiments, the individual has symptoms of or has been diagnosed with amyotrophic lateral sclerosis. The MSP may be administered to the patient intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intrapleurally, intranasally, intravitreally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularly, mucosally, intrapericardially, orally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, or via a lavage, for example.
40530616.1 [0010] In some embodiments, there is a pharmaceutical composition for treating ALS, comprising a therapeutically effective amount of MSP. In specific embodiments, the MSP comprises or is the amino-terminal 125 amino acid domain of VapB protein. In specific cases, the composition is a fragment, derivative, shuffling product, conjugate, or any modified product of MSP. In other embodiments, the more than the MSP domain of VAPB is utilized in methods and/or compositions of the disclosure; in some cases, most or all of VAPB is employed in the methods and/or compositions. An example of VAPB nucleotide sequence is at GenBank® Accession No. AY358464; an example of VAPB amino acid sequence is at GenBank®
Accession No. AAQ88829, both of which are incorporated by reference herein in their entirety.
[0011] In specific embodiments, there is use of a MSP composition for treating
ALS.
[0012] In certain embodiments, there is a pharmaceutical composition for treating ALS, comprising a therapeutically effective amount of a molecule that binds to, activates, or inhibits EphA4, Robo, and/or Dlar receptors (or human equivalents thereof).
[0013] In some embodiments, there is a pharmaceutical composition for treating ALS, comprising an amino-terminal domain of VapB protein that is greater than or less than 125 amino acids.
[0014] In some aspects, there is a pharmaceutical composition for treating ALS, comprising any domain of VapB protein that activates, binds to, or inhibits EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
[0015] In some embodiments, a VAPB paralog referred to as VAPA, may be employed in the disclosure (see, for example, SEQ ID NO:25 as the human VAPA sequence with SEQ ID NO:26 as its respective MSP domain). The VAPA protein has a MSP domain very similar to the VAPB MSP domain, and in particular embodiments the two MSP domains are functionally equivalent. Thus, in specific embodiments, there are compositions and methods relating to the use of SEQ ID NO:26 or functional derivates of fragments thereof, for the treatment of a neurological disorder, such as amyotrophic lateral sclerosis. In some aspects, compositions for treating ALS comprise a therapeutically effective amount of SEQ ID NO:26 or
40530616.1 functional derivates of fragments thereof. In particular embodiments, a functionally active derivative of MSP is provided to the individual, such as one that has one or more alterations compared to SEQ ID NO:26; in specific embodiments, there are one, two, three, four, five, six, seven, eight, nine, or ten or more alterations in the MSP derivative compared to SEQ ID NO:26. In particular embodiments, the MSP derivative lacks part of its N-terminus and/or C-terminus. In some cases, one or more internal amino acids are lacking compared to SEQ ID NO:26.
[0016] In embodiments of the disclosure, there is a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of a Major Sperm Protein (MSP) composition or functionally active fragment or derivative thereof to the individual. In specific embodiments, the neurological disorder is amyotrophic lateral sclerosis (ALS), including familial or sporadic. In specific cases, the individual has at least one symptom of ALS. In some cases, the individual is suspected of having ALS. In particular embodiments, the method further comprises the step of diagnosing ALS in the individual. In certain aspects, the MSP composition is provided to the individual multiple times. The MSP composition may be administered to the individual intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intrapleurally, intranasally, intravitreally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularly, mucosally, intrapericardially, orally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, or via a lavage. In some embodiments, the composition is provided in proteinaceous form, although it may be provided in nucleic acid form (including in an expression vector, such as a viral vector of any kind). In particular embodiments, the individual is provided an additional therapy for the neurological disorder or therapy for at least one symptom thereof. In specific embodiments, the neurological disorder is ALS. In some cases, the additional therapy is riluzole.
[0017] In embodiments of the disclosure, there is a pharmaceutical composition for treating ALS, comprising MSP or functionally active fragment or derivative thereof in a pharmaceutically acceptable excipient.
[0018] In embodiments of the disclosure there is a kit, comprising a pharmaceutical composition as disclosed herein, said composition housed in a suitable container. In some
40530616.1 embodiments, the kit further comprises an additional therapy for a neurological disorder or therapy for at least one symptom thereof.
[0019] In embodiments of the disclosure, there is a pharmaceutical composition comprising a MSP composition or functionally active fragment or derivative thereof. In specific embodiments, the composition comprises one or more modifications. In some cases, the one or more modifications extend the half life of the composition. In particular embodiments, the MSP composition comprises one or more polyethylene glycol groups; one or more immunoglobulins; at least one D amino acid; and/or a label, tag, or both. In some cases, the MSP composition is fused in-frame with another polypeptide. In certain embodiments, the functionally active fragment or derivative thereof is at least 80%, 85%, 90%, 95%, 97%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26. In some cases, the functionally active fragment or derivative thereof comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid alterations compared to SEQ ID NO:2 or SEQ ID NO:26. An alteration may comprise an amino acid substitution, deletion, addition, or inversion. In specific embodiments, a functionally active fragment or derivative thereof is no more than 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40 35, 30, or 25 amino acids in length. In certain cases, a functionally active fragment or derivative thereof comprises at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 120 contiguous amino acids of SEQ ID NO:2 or SEQ ID NO:26. In some embodiments, the functionally active fragment or derivative thereof comprises a N-terminal truncation. A N- terminal truncation may comprise absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26. In certain embodiments, a functionally active fragment or derivative thereof comprises a C-terminal truncation, such as a C-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26. In particular cases, a functionally active fragment or derivative thereof comprises a N-terminal truncation and a C-terminal truncation.
40530616.1 [0020] In embodiments, a MSP composition comprises an antibody fragment, such as Fc. In some embodiments, a MSP composition comprises collagen or albumin or both.
[0021] In some embodiments, a MSP composition is further defined as a MSP multimer or a MSP dimer or a MSP trimer or a MSP oligomer. In specific embodiments, a MSP composition is further defined as a peptide shuffled MSP molecule. In particular cases, a MSP composition comprises a MSP multimer comprsing at least two, three, four, or more MSP monomers.
[0022] In certain aspects of the disclosure, a composition comprises a collagen triple helix structure with MSP such that there is a trimer of MSP. In specific embodiments, MSP is utilized in a composition with a Fc as a monomer or dimer.
[0023] In embodiments of the disclosure, there is a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of FOXOl in the individual. In particular cases, the agent is a small molecule, nucleic acid, peptide, or protein. A nucleic acid may be a polynucleotide that encodes at least part of FOXOl. In some embodiments, a method further comprises the step of providing a therapeutically effective amount of a MSP composition or functionally active fragment or derivative thereof to the individual.
[0024] In embodiments of the disclosure, there is a method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of Osbp in the individual. In some cases, the agent is a small molecule, nucleic acid, peptide, or protein. A nucleic acid may be a polynucleotide that encodes at least part of Osbp. In certain aspects, a method further comprises the step of providing a
therapeutically effective amount of a MSP composition or functionally active fragment or derivative thereof to the individual.
40530616.1 Other neurological disorders that may be treated with methods and/or compositions of the disclosure include at least Alzheimer's disease; autism; cerebral palsy, dyslexia; Huntington's; multiple sclerosis; Parkinson's disease; and so forth.
BRIEF DESCRIPTION OF THE DRAWINGS
[0025] For a more complete understanding of the present disclosure, reference is now made to the following descriptions taken in conjunction with the accompanying drawings, in which:
[0026] FIG. 1A is a western blot of His-MSP eluates from Nickel affinity chromatography.
[0027] FIG. IB is a western blot of His-MSP before and after dialysis.
[0028] FIG. 1C is a western blot of His-MSP after incubation at 37 0 C for the indicated periods of time.
[0029] FIG. 2 displays MSP as a domain of VapB, and as a free peptide after cleavage from VapB, where it binds to EphA4, Robo, and Dlar receptors.
[0030] FIG. 3 shows localization of VapB to the ER and cell membrane.
[0031] FIG. 4 shows that P58S mutant protein accumulates in cytoplasmic inclusions.
[0032] FIG. 5 shows where the MSP domain is cleaved from the full length VapB protein.
[0033] FIG. 6A is a graph that shows that injection of His-MSP prolongs the survival of SODl G93A transgenic mice over control mice (treated with PBS).
[0034] FIG. 6B is a graph that shows the average survival days of His-MSP-treated SODl G93A transgenic mice was longer than control mice (treated with PBS).
40530616.1 [0035] FIG. 7 shows that injection of His-MSP rescues the motor function of SOD1 G93A transgenic mice in a rotarod assay.
[0036] FIG. 8 shows that fat levels in body wall muscle of wild- type and vpr-1 mutant worms. (A) DIC images of muscle in live adult hermaphrodites. Arrowheads indicate lipid-like droplets. Bar, 5 μιη. (B) Transmission electron micrographs of body wall muscle cytoplasm in wild-type and vpr-l(tml411 ) mutant hermaphrodites. Light blue color demarcates muscle boundary. L, Lipid-like droplet. Bar, 0.5 μιη. (C) Fluorescent images of muscle in live adult hermaphrodites fed Bodipy-FAs. Closeup images of boxed areas are shown below.
Arrowheads indicate examples of Bodipy- FA-stained droplets. Anterior is to the left in all panels. Bars, 50 μιη. (D) High magnification images of muscle showing Bodipy-FA-stained fluorescent droplets and droplets observed by DIC microscopy. Bar, 5 μιη. (E) Comparison of total ion chromatograms of wild-type and vpr-l(tml411 ) mutant adults extracts for 18:0 TAG (Neutral Loss 284) and phosphatidylethanolamine (Neutral Loss 141).
[0037] FIG. 9 provides vpr-1 mosaic analysis. (A) Analysis of vpr-1 genetic mosaics showing the lineages of major tissues. Each circle indicates one genetic mosaic worm. Points at which the genomic copy of vpr-1 (+) was lost and the resulting phenotype are shown. (B) Representative DIC images of muscle in vpr-l(tml411 ) mutant mosaic worms. Ex vpr-1 (+) indicates expression of the vpr-1 genomic locus via an extrachromosomal array. Arrowheads indicate fat droplets. Bar, 5 μιη.
[0038] FIG. 10 shows the effect of tissue-specific vpr-1 expression on fat levels. (A) DIC images of muscle in live wild- type and vpr-1 (tml411) mutant hermaphrodites expressing wild-type VPR-1 or VPR-1 (P56S) under indicated tissue-specific promoters.
Arrowheads indicate lipid-like droplets. Bar, 5 μιη. (B) Sudan Black B staining images of i mutants expressing vpr-1 under the unc-119 pan-neuronal promoter. Arrows indicate muscle fat droplets. Anterior is to the left in all panels. Wild- type controls (FIG. 17) are similar to transgenic vpr-l(tml411 ) mutants expressing uncll9p::vpr-l. Low magnification bars, 50 μιη; high magnification bars, 25 μιη.
40530616.1 [0039] FIG. 11 demonstrates effect of Arp2/3 inactivation on muscle fat levels. DIC and fluorescent images of muscle in live 3-day-old hermaphrodite worms fed Bodipy-FAs. arx-2 encodes the Arp2 component of the Arp2/3 complex. Arrowheads indicate Bodipy-FA- stained fat droplets. Bar, 5 μιη.
[0040] FIG. 12 provides DAF-16 activity in vpr-1 mutants. (A) DIC and fluorescent images of muscle in live 3-day-old hermaphrodite worms fed Bodipy-FAs.
Arrowheads indicate Bodipy-FA-stained droplets. Wild-type controls (not shown) are similar to daf-16(mu86) mutants (See figures 1C and 4). Bar, 5 μιη. (B) Pharyngeal pumping rates of 1 -day- old adult hermaphrodites. Wild type (236.7+21.1 [n=l 1]) and vpr-1 (tml411 ) mutants (220.1+9.6 [n=l 1]) have similar pharyngeal pumping rate. Error bars represent SD. *P > 0.05 compared to wild type. **P > 0.05 compared to vpr-l(tml411) mutant. (C) Transgenic worms expressing GFP under control of the sod-3 promoter, a direct DAF-16/FoxO target. Anterior is to the left in all panels. Bar, 50 μιη.
[0041] FIG. 13 shows DAF-16 localization and activity in wild- type and mutant worms. (A) Transgenic strains expressing DAF-16::GFP under its endogenous promoter.
Transgenic controls raised at 20°C are similar to those raised at 20°C then shifted to 35°C for 30 minutes (see panel B for quantification). Close up images of boxed areas are shown. Anterior is to the left in all panels. Low magnification bar, 50 μιη; high magnification bar, 25 μιη. (B) Quantification of DAF-16::GFP localization in control (n=157) and vpr-l(tml411) mutants (n=49). (-), incubation under normal growth condition; (+), incubation at 35°C for 30 minutes. (C) Magnified images showing transgenic lines expressing GFP under the sod-3 promoter, arx-2 encodes Arp2. Arrows indicate vulva muscle region. Anterior is to the left in all panels. Bar, 50 μιη. (D) Average sod-3p::GFP fluorescent intensity of the intestine (N > 10 worms). Error bars represent SD. *P < 0.05. (E) sod-3p::GFP in vpr-1 mutant hermaphrodites in the presence or absence of wild-type (WT) sperm. Anterior is to the left in all panels. Bar, 50 μιη.
[0042] FIG. 14 provides the effect of DAF-16 inactivation on muscle mitochondria.
(A) Muscle mitochondrial tubules in indicated genotypes visualized using mitochondrial matrix- targeted GFP (mitoGFP). Arrowheads indicate fat droplets. Asterisks indicate nucleus. Bar, 5 μιη.
(B) MitoTracker CMXRos staining of wild-type and mutant muscle. Asterisks indicate nucleus.
40530616.1 Bar, 5 μηι. (C and D) Oxygen consumption rates of wild-type and mutant hermaphrodites.
Measured consumption rates were normalized by protein content (C) or number of worms (D). Error bars represent SD. *, P < 0.001 compared to wild type. Oxygen consumption rate of wild- type and vpr-l(tml411 ) mutants includes published data [15] measured together with vpr- l(tml411) daf-16(mu86) mutants.
[0043] FIG. 15 shows the effect of DAF-16 inactivation on ATP level and lifespan. (A) ATP concentration in wild-type and vpr-l(tml411) mutant adult extracts. *, P <0.001 compared to wild type. Error bars represent SD. ATP concentration of wild-type and vpr- l(tml411) mutants at 1-day-old adults include published data (Han et ah, 2012) measured together with vpr-l(tml411) daf-16(mu86) mutants. (B) Lifespan measurements of indicated genotypes. The lifespan of daf-16(mu86) mutants was similar to the wild type, as previously shown.
[0044] FIG. 16 demonstrates effect of Vapb ablation on fasting/refeeding energy metabolism in mice. (A) TAG concentration in GA muscle and liver of wild-type (+/+) and Vapb knock-out (-/-) mice after 24-hour fasting (red) or 24 hours fasting followed by 6 hours of refeeding (blue). (B and C) Quantitative RT-PCR of indicated genes in liver (B) and TA muscle (C) of wild-type (+/+) and Vapb knock-out (-/-) mice after 24-hour fasting (red) or 24 hours fasting followed by 6 hours of refeeding (blue). Relative mRNA levels are shown on the Y-axis. #, P < 0.05 compared to fed mice of the same genotype. *, P < 0.05 compared to +/+ under the same condition.
[0045] FIG. 17 shows Sudan Black B staining in wild type and vpr-1 mutants. 1- day-old adult wild- type and vpr-1 (tml411 ) hermaphrodites were stained using Sudan Black B. Arrowheads indicate fat droplets in body wall muscle. Anterior is to the left in all panels. Boxed regions are magnified 5X below. Low magnification bars, 50 μιη; high magnification bars, 10 μτη.
[0046] FIG. 18 provides ER stress assays in wild-type and vpr-1 mutant worms. (A) Integrated transgenic lines expressing GFP under the hsp-4 promoter (hsp-4p::GFP) with and without tunicamycin treatment, which induces ER stress. Anterior is to the left in all panels. Bar,
40530616.1 5 mm. (B) Tunicamycin sensitivity in wild-type and vpr-l(tml411) mutants hermaphrodites. Y- axis indicates the percentage of worms that developed to the adult stage in the presence of 5 μg/ml tunicamycin. Three independent measurements were performed.
[0047] FIG. 19 demonstrates effect of sperm presence on muscle fat droplets in vpr-1 mutants. DIC and fluorescent images of muscle in live 3-day-old vpr-1 mutant
hermaphrodite worms fed Bodipy-FAs. Mating with wild type (WT) males provides sperm into the uterus. Anterior is to the left in all panels. Arrowheads indicate lipid-like droplets. Bar, 50 μιη.
[0048] FIG. 20 shows loss of Vap causes accumulation of membrane proteins in the adult neurons. (A-B) Neurons in the adult brain of 1 day-old adult flies stained with anti- Chaoptin and anti-Elav antibodies. In control flies, Chaoptin labels individual synapses in the lamina (dots) as previously published (Hiesinger et ah, 2005). However, it is also expressed in few cortical neurons of the brain at low levels (A, arrows). In vap null mutant brains, Chaoptin accumulates in the cytoplasm of the neurons of the cortex (B, B' and B", arrows). A precise excision of P{Mae-UAS.6.11 J in vap was used as a control in A-A" (see method). (C-F) Staining of cortical neurons in the adult brain of 1 day-old flies with anti-Robo-1 and anti-Elav antibodies (C, D) and N-Cadherin (E, F). In vap null mutants, Robo-l(D) and N-Cadherin (F) accumulates in the cytoplasm of a subset of neurons (arrows). Op: optic lobe.
[0049] FIG. 21 shows loss of Vap causes accumulation of membrane proteins in the ER of neurons. (A) Staining of affected neurons in the adult brain of the vap null mutant. Accumulated Chaoptin protein is surrounded by BOCA, the ER marker (indicated by * in A"). (B) Staining of cortical neurons in the adult brain of 1 day-old flies expressing the membrane anchored GFP (CD8-GFP) with anti-Chaoptin and anti-GFP antibody. In the vap null mutant neurons, some neurons accumulate both CD8-GFP (arrows, B') and Chaoptin (arrows, B").
[0050] FIG. 22 demonstrates that loss of Vap causes an aberrant ER expansion. (A- D) TEM analysis of neurons of control (A) and of vap null mutants (B-D). Large cytoplasmic vacuoles in the neurons of the vap null mutant (asterisk (*), B). The vacuole is contiguous with the nuclear membrane (arrow heads, C). The vacuole is also decorated with electron-dense
40530616.1 ribosomes (arrows, D), suggesting that the vacuoles are aberrantly expanded ER. N: Nucleus. A precise excision line was used as a control.
[0051] FIG. 23 demonstrates that loss of VAP causes ER stress. (A-D) An ER stress reporter Xbpl-GFP is significantly upregulated in the neurons in the adult brain of 1 dayold the vap null mutant (B and D), but not in the control flies (A and C). A precise excision line was used as a control. (E-F) Neurons of 1 day-old adult brain stained with anti-Bip, an ER stress marker. Bip is upregulated in the neurons of the vap null mutant (arrows in F). (G-H) Staining of the neurons in the cortex of the adult brain of control (G) and the vap null mutant (H).
Ubiquitinated proteins accumulate in the cytoplasm of cortical neurons in adult vap null mutant (H and FT), but not in control adults (G and G').
[0052] FIG. 24 shows that Osbp colocalizes with Vap in the ER. (A) Vap and Osbp interaction in a GST pull-down assay. GST- Vap WT binds Osbp, but GST-VapALS8 binding is severely reduced. GST- Vap WT and GST-VapALS8 were expressed in bacteria and purified with glutathione sepharose beads. GST- Vap proteins linked to beads were incubated with lysate extracted from Schneider's S2 cells expressing Osbp. The precipitate and input were analyzed with immunoblots using anti-Osbp antibody and anti-GST antibody. (B) Immunoblot analysis of proteins extracted from flies overexpressing Osbp, control (WT) and Aosbp over deficiency Df(3R)ED622, AosbplDf flies. Note the lack of Osbp protein expression in AosbplDf, showing the specificity of the Osbp antibody (GP89). (C) MARCM analysis showing the specificity of guinea pig anti-Osbp (GP89) antibody. A portion of the wing imaginal disc stained with anti-Osbp antibody (GP89). GFP marks the osbp null mutant cells. (D) Immuno staining of a WT salivary gland cell with anti-Osbp (GP89) and anti-Vap antibody (Rb92). Osbp colocalizes with Vap in the ER.
[0053] FIG. 25 shows that Osbp is mislocalized to the Golgi in vap null mutant cells. (A-B) MARCM analysis shows that Osbp accumulates in cytoplasmic punctae of vap null mutant cells (B and arrows in B') but not in wild type cells (A and A'). (C-D) Osbp accumulates in the Golgi of vap null mutant cells. Immuno staining of the 1 day-old adult brain of WT
(control) (C) and vap null mutant neurons (D) with anti-Osbp (C and D') and anti-Msl20 (C" and D") antibody, a Golgi marker. Osbp barely colocalizes with the Golgi marker Ms 120 KDa
40530616.1 protein in the WT control (C-C"), whereas Osbp colocalizes or is closely associated with Msl20 KDa protein in vap null mutant neurons (arrows, D-D").
[0054] FIG. 26 shows that ALS8 mutation causes a partial loss of function of VAP. (A) Comparable levels of expression of vapWT and vapALS8 transgenes. Immunoblot analysis of proteins extracted from WT (control) and transgenic flies carrying genomic vapWT (VK31) and vapALS8 (VK31) in the vap null mutant background with anti-Vap and anti-Actin antibodies. (B) Longevity of adult flies. The vapALS8 transgene can rescue the lethality associated with loss of Vap, but VapALS8 is not as active as VapWT. An additional copy of vapWT can compensate for the defects associated with vapALS8. Genotypes shown: Δναρ (vap null mutant), Δναρ; vapWT (vap null mutant carrying genomic vapWT, line F7 and line VK31), J vap; vapALS8 (vap null mutant carrying genomic vapALS8 , line M6 and line VK31), Δναρ; vapWT/ vap ALS8 (vap null mutant carrying genomic vapWT and vapALSS). Line F7 and M6 are P-element mediated transgenes. Line VK31 is site specific integrated transgene in VK31. Longevity: Δναρ (pharate lethal), control (64 days), Δναρ; vapWT (F7) (52 days), Δναρ; vapWT (VK31) (62 days), Δναρ; vapALS8 (M6) (23 days, * P<0.001 compared to Δναρ; vapWT(Vl) Δναρ; vapALS8 (VK31) (14 days, * P<0.001 compared to Δναρ; vapWT (VK31)), Δναρ; vapWT(F7)/ vap ALS8 (M6) (65days) and Δναρ; vapWT/ vap ALS8 (VK31) (67days). Error bars represent STD. (C) Longevity of adult flies. The vapALS8 transgenes rescue the lethality associated with loss of vap, but VapALS8 is not as active as VapWT. Genotypes: Avap; vapWT (vap null mutant carrying genomic vapWT, line Ml 1, F6 and F10). Δναρ; vapALS8 (vap null mutant carrying genomic vapALS8 , line V37, V38, V40, V42, V43, V45, and V46). All lines carry P-element mediated transgenes. Error bar represents STD. (D) Flight test. vapWT can suppress the flight defects associated with vapALS8. Δναρ; vapALS8 (VK31) flies exhibit flight defects at day 12 (* P<0.001 compared to control), but not at day 4 after eclosion. However, Δναρ; vapWT/ vapALS8 flies do not show flight defects. Flies were individually dropped into a plastic cylinder, and the height at which they landed was recorded. The shorter the distance from the bottom to their landing point, the worse their ability to fly. Error bar represent SEM.
[0055] FIG. 27 shows that ALS8 mutation causes progressive defects in adult fly brain. (A) VapALS8 causes adult brain degeneration. Brain section of 12-day-old adult flies.
40530616.1 Δναρ; vapALS8 (VK31), but not Avap; vapWT (VK31) flies show vacuolation of brains in the optic lobe and central lobe. An additional copy of vapWT suppresses the defects associated with VapALS8. op: optic lobe, cen: central lobe. (B) Loss of Vap causes functional defects in motor neurons in adult flies. In the vap null mutant one day old flies, the TTM muscles are unable to follow a 200Hz stimulus but they can almost respond properly when stimulated at 10 Hz. (B) shows the difference between WT control (n=16) and vap null mutants (n = 9) at the 20th pulse when stimulated at 10Hz (control p =l; Avap p= 0.76 + 0.15; mean + SEM), 20Hz (control p =1; Avap p = 0.49 + 0.16), 50Hz (control p = 1; Avap p= 0.23 + 0.12), 100Hz (control p=l; Avap p = 0.14 + 0.03), 200Hz (control p = 0.97 + 0.02; Avap p = 0.04 + 0.03). n: the number of flies used. * p<0.05 and ** p<0.005. (C) VapALS8 causes functional defects in motor neurons in adult flies. Comparison of 6 day old and 12 day old flies that are responding to high frequency nerve stimulation (200Hz) between control, Avap; vapWT (VK31) and Avap; vapALS8 (VK31) at 20th pulse. There is no difference in 6 day and 12 day old control flies (6 day p = 0.99 + 0.013, n=8; 14 day: p = 0.98+0.025, n = 9) as well as Avap; vapWT (6 day: 0.98 + 0.017, n = 8; 12 day: 0.99 + 0.010, n = 12). However, there is a significant reduction in the ability to follow a 200Hz stimulus in Avap; vapALS8 (6 day: p = 0.87 + 0.057, n = 17; 12 day: p = 0.57 + 0.116, n = 15. Controls are flies that have a precise excision of P{Mae-UAS.6.11 J.
[0056] FIG. 28 shows that expression of human OsbpL8 suppresses the ER defects associated with loss of Vap. (A-F, H) Immuno staining of the adult brain of WT (control) (A, B), vap null mutant (C, D), vap null mutants expressing HA-hOSBPL8 (E, F) and the vap
heterozygous mutant expressing HA-hOSBPL8 with neuronal GAL4 (D42) driver (H).
Expression of human OsbpL8 suppresses the upregulation of Bip and accumulation of
Ubiquitinated proteins caused by loss of Vap (compare E and F to C and D). (G) Graphical overview of the Osbp and hOSBPL8 proteins. Human OsbpL8 contains a transmembrane domain, but not a VAP binding FFAT motif. FFAT: VAP binding site, ORD: OSBP related domain, TM: transmembrane domain.
[0057] FIG. 29 demonstrates that MSP treatment induces phosphorylation of RTKs. A. MSP stimulation activates protein phosphorylation signaling in HEK293 cell. HEK293 cells were serum starved for 16hrs before stimulated with His-MSP (lOOng/ml). Cells were
40530616.1 harvested in lysis buffer and blotted with anti-phosphor- tyrosine antibody. B. Protein phosphorylated microarray analysis reveals changes of protein tyrosine phosphorylation upon MSP treatment. Cell lysate from Fig. lA were tested by PathScan RTK signaling antibody array kit. The level of phosphorylation is reflected by the signal intensity of each dot. C. Heat map of the intensity measured in Fig. IB. D. Predicted MSP downstream signaling network (using STRING 9.05).
[0058] FIG. 30 indicates that MSP treatment increases the number of primary hippocampal neuron synapses in vitro. A. Confocal microscopy images of MSP or PBS treated hippocampal neuron cultures. Cells are stained with the synaptic marker Bassoon, dendritic marker Map2, and nuclear marker DAPI (scale bar 50 μιη). B. Quantification of number of Bassoon+ synapses on hippocampal neuron dendrites of cultures treated with either MSP (100 ng/ml) or PBS. Each bar indicates a separate culture. Numbers of synapses on 250 μιη of dendritic length of 10 fields of view of separate population of neurons were quantified. MSP treated neurons showed significantly more synapses than PBS controls. n= 10 fields of view for each culture, * p<0.05.
[0059] FIG. 31 illustrates an example of a VAPB expression construct for utilization with recombinant adeno-associated virus 8 in an ALS model, SOD1-G93A. Animals are injected intracerebroventricularly with AAV8-VAPB at birth to produce widespread expression of the viral construct in neurons throughout the brain and into the spinal cord.
[0060] FIG. 32 provides an example of a scheme for assessing the efficacy of VAPB treatment delivered by intracranial AAV8 injection in SOD mice .Wild-type and SOD1 transgenic mice were injected with AAV8-VAPB at postnatal day 0, then tested for behavioral symptoms from postnatal day 80 until death. Behavioral readouts include motor performance on the rotorod assay and an overall symptom assessment based on the scale from Vercelli et al (2008). Tissue is harvested from brain, spinal cord, blood, heart, kidney, and muscle at endpoint for analysis of viral expression.
[0061] FIG. 33 illustrates that AAV8-VAPB treatment delays the loss of motor ability during early stages of disease. SOD1 mice were administered AAV8-VAPB at birth and
40530616.1 tested by rotarod from postnatal day 80 until they were no longer able to perform the task, approximately postnatal day 150. The findings show significant improvements in the VAPB- treated SOD mice prior to postnatal day 120.
[0062] FIG. 34 demonstrates survival rate for SOD1 transgenic mice treated with VAPB-AAV8 at birth. There is a small improvement in survival with VAPB treatment.
[0063] FIG. 35 shows that the initial decline in gross motor symptoms is delayed by AAV8-VAPB treatment of SOD1 transgenic mice compared to untreated SOD animals.
[0064] FIG. 36 demonstrates that there is strong overexpression of VAPB protein in the brains of wild- type and SOD1 transgenic mice treated at birth with AAV8-VAPB.
[0065] FIG. 37 demonstrates increased levels of MSP fragment in the plasma of wild-type mice treated with AAV8-VAPB at birth.
DETAILED DESCRIPTION
[0066] As used herein the specification, "a" or "an" may mean one or more. As used herein in the claim(s), when used in conjunction with the word "comprising", the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a second or more. Still further, the terms "having", "including", "containing" and "comprising" are interchangeable and one of skill in the art is cognizant that these terms are open ended terms. Some embodiments of the disclosure may consist of or consist essentially of one or more elements, method steps, and/or methods of the disclosure. It is contemplated that any method or composition described herein can be implemented with respect to any other method or
composition described herein.
I. General Embodiments
[0067] General embodiments provided herein concern methods and compositions for treating one or more neurological disorders, such as ALS, with part or all of the Major Sperm Protein (MSP) domain of VapB. The individual that is treated shows at least one neurological symptom associated with ALS or has at least one risk factor to delay or prevent the onset of ALS.
40530616.1 An exemplary risk factor is a family member with ALS and/or having one or more mutations linked to ALS.
[0068] Data presented herein implicates the loss of MSP as a key initiator of disease progression. Therefore, delivery of part or all of an effective amount of MSP or a derivative thereof to an individual, including into the blood, or by other means of peripheral delivery, for example, provides a beneficial therapeutic intervention for ALS, including sporadic and familial ALS cases.
II. Amyotrophic Lateral Sclerosis (ALS)
[0069] Amyotrophic lateral sclerosis (ALS), which is also referred to as Lou Gehrig's disease, is a rapidly progressive, fatal neurological disease that attacks the nerve cells that control voluntary muscles, referred to motor neurons. In ALS, both the upper motor neurons (in the brain) and the lower motor neurons (in the spinal cord) degenerate or die and no longer send messages to muscles, and the muscles gradually weaken and then atrophy. Ultimately, all muscles under voluntary control are affected, and individuals lose their strength and the ability to move their arms, legs, and body. Most people with ALS die from respiratory failure when muscles in the diaphragm and chest wall fail, losing the ability to breathe without a ventilator.
[0070] Symptoms of ALS may be overlooked initially, given their subtle nature. The earliest symptoms may include fasciculations, cramps, tight and stiff muscles (spasticity), muscle weakness affecting an arm or a leg, slurred and nasal speech, and/or difficulty chewing or swallowing. To be diagnosed with ALS, individuals need to have signs and symptoms of both upper and lower motor neuron damage that cannot be attributed to other causes. Thus, diagnosis of ALS is based on the symptoms and signs observed by a medical provider and a series of tests to rule out other diseases. Medical providers may assess over time whether symptoms such as muscle weakness, atrophy of muscles, hyperreflexia, and spasticity are increasing in their severity.
[0071] Because ALS symptoms at the onset can be similar to those of a wide variety of other, more treatable diseases or disorders, certain tests may be performed to exclude the other conditions. Such tests include electromyography (EMG), a special recording technique
40530616.1 that detects electrical activity in muscles or a nerve conduction study that measures electrical energy by assessing the nerve's ability to send a signal. In some cases, the medical provider may utilize magnetic resonance imaging (MRI) of the brain and/or spinal cord to exclude certain conditions.
[0072] Based on the person's symptoms and findings from the examination and from these tests, the physician may order tests on blood and urine samples to eliminate the possibility of other diseases as well as routine laboratory tests.
[0073] In some cases, riluzole is provided to the individual for therapy. In specific embodiments of the disclosure, riluzole or another ALS treatment is provided in addition to compositions of the disclosure. Other medications that may be employed with the disclosure include those that alleviate certain symptoms, such as those that help reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, painkillers, depression treatment, treatment for sleep disturbances, and/or constipation medication.
III. MSP Proteins and Derivatives Thereof
[0074] Embodiments include the delivery of part or all of MSP to an individual in need thereof. The individual may be deficient in MSP level. The individual may be known to have a neurological disorder or suspected of having a neurological disorder or at risk for having a neurological disorder, such as ALS.
[0075] In some embodiments, the entire MSP is provided to an individual. In some cases, a functionally active fragment or derivative of MSP is provided to the individual. In certain embodiments, only part of the entire MSP is provided to the individual, such as one having no more than 123, 122, 121, 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, 15, or 10 amino acids of SEQ ID NO:2 or SEQ ID NO:26. In some embodiments, a particular domain of MSP is included in the MSP derivative. In specific embodiments, the MSP fragment or derivative has at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120 amino acids of SEQ ID NO:2 or SEQ ID NO:26. In particular embodiments, the MSP derivative is modified compared to SEQ ID NO:2 or SEQ ID NO:26, such as having 1, 2, 3, 4, ,5 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more
40530616.1 alterations compared to SEQ ID NO:2 or SEQ ID NO:26, and the alteration may include a deletion, substitution, inversion, and so forth.
[0076] In particular aspects of the disclosure, the MSP derivative is at least 70%, 75%, 80%, 82%, 85%, 87%, 90%, 92%, 95%, 97%, 98%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26. In certain aspects the MSP composition will have all or part of the amino acid sequence of SEQ ID NO:2 or SEQ ID NO:26. The MSP compositions described herein may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more variant amino acids within at least, or at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, or more contiguous amino acids, or any range derivable therein, of SEQ ID NO:2 or SEQ ID NO:26.
[0077] Thus, as modifications may be made in the structure of the proteins, while obtaining molecules having similar or improved characteristics, such biologically functional equivalents are also encompassed within the present disclosure.
[0078] Although in particular embodiments MSP or fragment or derivative thereof is delivered to an individual in proteinaceous form, in alternative embodiments a nucleic acid encoding the MSP or fragment or derivative thereof is delivered to the individual.
In some embodiments, fragment refers to a domain of VapB that is smaller or larger than MSP that still retains the biological activity of MSP. As used herein, derivatives refers to compounds that are derived from chemical modifications to a starting compound. As used herein, shuffling product is a peptide in which an amino acid, amino acids, or groups of amino acids have been transposed from their original sequence. As used herein, a conjugate is molecule that has a molecule, functional group, polymer, protein or nanoparticle attached to it. As used herein, modified product is a molecule which has been chemically altered.
40530616.1 [0079] In some aspects, there are compositions comprising a fragment, derivative, shuffling product, conjugate or any modified product of MSP. In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising a therapeutically effective amount of a molecule that binds to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof). In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising a therapeutically effective amount of a molecule that activates EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof). In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising a therapeutically effective amount of a molecule that inhibits EphA4, Robo, and/or Dlar receptors (and/or human
equivalents thereof). In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising an amino-terminal domain of VapB protein that is greater than 125 amino acids. . In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising an amino-terminal domain of VapB protein that is less than 125 amino acids. In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising any domain of VapB protein that activates to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof). In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising any shuffling product of VapB protein that binds to EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).. In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising any shuffling product of VapB protein that inhibits EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof). In some aspects, there are compositions for treating amyotrophic lateral sclerosis, comprising any shuffling product of VapB protein that activates EphA4, Robo, and/or Dlar receptors (and/or human equivalents thereof).
[0080] In embodiments of the disclosure, the MSP comprises one or more modifications. Although any suitable modifications are encompassed in the disclosure, in certain embodiments the one or more modifications extend the half life of the composition. In particular cases, the MSP composition comprises one or more polyethylene glycol groups, one or more immunoglobulins, at least one D amino acid, and/or a label, tag, or both.
40530616.1 [0081] In certain aspects, the MSP composition is fused in-frame with another polypeptide.
[0082] In particular embodiments, a functionally active fragment or derivative thereof is at least 80%, 85%, 90%, 95%, 97%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26. The functionally active fragment or derivative thereof may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid alterations compared to SEQ ID NO:2 or SEQ ID NO:26. An alteration may be of any kind, such as an amino acid substitution, deletion, addition, or inversion.
[0083] In some cases, a functionally active fragment or derivative thereof is no more than 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40 35, 30, or 25 amino acids in length. In certain cases, a functionally active fragment or derivative thereof comprises at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 120 contiguous amino acids of SEQ ID NO:2 or SEQ ID NO:26.
[0084] Particular functionally active fragments or derivatives thereof of MSP comprise a N-terminal truncation, such as a N-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
[0085] Certain functionally active fragments or derivatives thereof of MSP comprise a C-terminal truncation, such as a C-terminal truncation that comprises absence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
[0086] Certain MSP functionally active fragments or derivatives thereof comprise a N-terminal truncation and a C-terminal truncation.
[0087] Particular MSP compositions comprise an antibody, including an antibody fragment, such as Fc. In some cases, MSP comprises collagen or albumin.
40530616.1 [0088] Embodiments include a MSP-Fc composition, a MSP-albumin composition, MSP-collagen like scaffold molecules, and so forth, and in specific embodiments such compositions have longer pK and half-life in vivo.
[0089] In certain aspects, MSP binds to different receptors. In embodiments, a MSP composition is further defined as a MSP multimer, which may be a dimer, trimer, and so forth. A MSP multimer may be comprised of 1, 2, 3, 4, 5, or more MSP monomers. In specific embodiments, a MSP composition is an oligomer or a peptide shuffled MSP molecule.
A. Modified Polynucleotides and Polypeptides
[0090] The MSP functionally active derivative or fragment may be referred to as a biological functional equivalent, and it may comprise a polynucleotide that has been engineered to contain distinct sequences while at the same time retaining the capacity to encode the "wild- type" or standard protein. This can be accomplished to the degeneracy of the genetic code, i.e., the presence of multiple codons, which encode for the same amino acids. In one example, one of skill in the art may wish to introduce a restriction enzyme recognition sequence into a
polynucleotide while not disturbing the ability of that polynucleotide to encode a protein.
[0091] In another example, a polynucleotide may be (and encode) a biological functional equivalent with more significant changes. Certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies, binding sites on substrate molecules, receptors, and such like. So-called "conservative" changes do not disrupt the biological activity of the protein, as the structural change is not one that impinges of the protein's ability to carry out its designed function. However, in some cases the alteration is not a conservative substitution. It is thus contemplated by the inventors that various changes may be made in the sequence of genes and proteins disclosed herein, while still fulfilling the goals of the present disclosure.
[0092] In terms of functional equivalents, it is well understood by the skilled artisan that, inherent in the definition of a "biologically functional equivalent" protein and/or
polynucleotide, is the concept that there is a limit to the number of changes that may be made
40530616.1 within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity. Biologically functional equivalents are thus defined herein as those proteins (and polynucleotides) in selected amino acids (or codons) may be substituted. Functional activity includes the ability of the protein form to bind EphrinA4, Dlar, and/or Robo receptors (and/or human equivalents thereof). Furthermore, one could test for biological activity by microinjecting MSP equivalents into the C. elegans gonad. As shown in Miller et al. 2001 and Tsuda et al. 2008, microinjecting MSPs into the gonad stimulates oocyte maturation and muscle contraction, which can be quantified.
[0093] In general, the shorter the length of the molecule, the fewer changes that can be made within the molecule while retaining function. Longer domains may have an intermediate number of changes. The full-length protein will have the most tolerance for a larger number of changes. However, it must be appreciated that certain molecules or domains that are highly dependent upon their structure may tolerate little or no modification.
[0094] Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and/or the like. An analysis of the size, shape and/or type of the amino acid side-chain substituents reveals that arginine, lysine and/or histidine are all positively charged residues; that alanine, glycine and/or serine are all a similar size; and/or that phenylalanine, tryptophan and/or tyrosine all have a generally similar shape. Therefore, based upon these considerations, arginine, lysine and/or histidine; alanine, glycine and/or serine; and/or phenylalanine, tryptophan and/or tyrosine; are defined herein as biologically functional equivalents.
[0095] To effect more quantitative changes, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and/or charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and/or arginine (-4.5).
40530616.1 [0096] The importance of the hydropathic amino acid index in conferring interactive biological function on a protein is generally understood in the art (Kyte & Doolittle, 1982, incorporated herein by reference). It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index and/or score and/or still retain a similar biological activity. In making changes based upon the hydropathic index, the substitution of amino acids whose hydropathic indices are within +2 is preferred, those which are within +1 are particularly preferred, and/or those within +0.5 are even more particularly preferred.
[0097] It also is understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biological functional equivalent protein and/or peptide thereby created is intended for use in immunological embodiments, as in certain embodiments of the present disclosure. U.S. Patent 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and/or antigenicity, i.e., with a biological property of the protein.
[0098] As detailed in U.S. Patent 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 + 1); glutamate (+3.0 + 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 + 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4). In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within +2 is preferred, those which are within +1 are particularly preferred, and/or those within +0.5 are even more particularly preferred.
B. Altered Amino Acids
[0099] The present disclosure, in many aspects, relies on the synthesis of peptides and polypeptides in cyto, via transcription and translation of appropriate polynucleotides. These peptides and polypeptides will include the twenty "natural" amino acids, and post-translational modifications thereof. However, in vitro peptide synthesis permits the use of modified and/or unusual amino acids.
40530616.1 C. Mimetics
[0100] In addition to the biological functional equivalents discussed above, the present inventors also contemplate that structurally similar compounds may be formulated to mimic the key portions of peptide or polypeptides of the present disclosure. Such compounds, which may be termed peptidomimetics, may be used in the same manner as the peptides of the disclosure and, hence, also are functional equivalents.
[0101] Certain mimetics that mimic elements of protein secondary and tertiary structure are described in Johnson et al. (1993). The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and/or antigen. A peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule.
[0102] Some successful applications of the peptide mimetic concept have focused on mimetics of β-turns within proteins, which are known to be highly antigenic. Likely β-turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains.
[0103] Other approaches have focused on the use of small, multidisulfide- containing proteins as attractive structural templates for producing biologically active
conformations that mimic the binding sites of large proteins. Vita et al. (1998). A structural motif that appears to be evolutionarily conserved in certain toxins is small (30-40 amino acids), stable, and high permissive for mutation. This motif is composed of a beta sheet and an alpha helix bridged in the interior core by three disulfides.
[0104] Beta II turns have been mimicked successfully using cyclic L-pentapeptides and those with D-amino acids. Weisshoff et al. (1999). Also, Johannesson et al. (1999) report on bicyclic tripeptides with reverse turn inducing properties.
40530616.1 [0105] Methods for generating specific structures have been disclosed in the art. For example, alpha-helix mimetics are disclosed in U.S. Patents 5,446,128; 5,710,245; 5,840,833; and 5,859,184. These structures render the peptide or protein more thermally stable, also increase resistance to proteolytic degradation. Six, seven, eleven, twelve, thirteen and fourteen membered ring structures are disclosed.
[0106] Methods for generating conformationally restricted beta turns and beta bulges are described, for example, in U.S. Patents 5,440,013; 5,618,914; and 5,670,155. Beta- turns permit changed side substituents without having changes in corresponding backbone conformation, and have appropriate termini for incorporation into peptides by standard synthesis procedures. Other types of mimetic turns include reverse and gamma turns. Reverse turn mimetics are disclosed in U.S. Patents 5,475,085 and 5,929,237, and gamma turn mimetics are described in U.S. Patents 5,672,681 and 5,674,976.
IV. Polynucleotides Encoding MSP
[0107] The present disclosure also encompasses a composition comprising a nucleic acid sequence encoding MSP domain as defined herein and cells harboring the nucleic acid sequence. The nucleic acid molecule is a recombinant nucleic acid molecule, in particular aspects and may be synthetic. It may comprise DNA, RNA as well as PNA (peptide nucleic acid) and it may be a hybrid thereof.
[0108] Furthermore, it is envisaged for further purposes that nucleic acid molecules may contain, for example, thioester bonds and/or nucleotide analogues. The modifications may be useful for the stabilization of the nucleic acid molecule against endo- and/or exonucleases in the cell. The nucleic acid molecules may be transcribed by an appropriate vector comprising a chimeric gene that allows for the transcription of said nucleic acid molecule in the cell. In this respect, it is also to be understood that such polynucleotides can be used for "gene targeting" or "gene therapeutic" approaches. In another embodiment the nucleic acid molecules are labeled. Methods for the detection of nucleic acids are well known in the art, e.g., Southern and Northern blotting, PCR or primer extension. This embodiment may be useful for screening methods for verifying successful introduction of the nucleic acid molecules described above during gene therapy approaches.
40530616.1 [0109] The nucleic acid molecule(s) may be a recombinantly produced chimeric nucleic acid molecule comprising any of the aforementioned nucleic acid molecules either alone or in combination. In specific aspects, the nucleic acid molecule is part of a vector.
[0110] The present disclosure therefore also relates to a composition comprising a vector comprising the nucleic acid molecule described in the present disclosure.
[0111] Many suitable vectors are known to those skilled in molecular biology, the choice of which would depend on the function desired and include plasmids, cosmids, viruses, bacteriophages and other vectors used conventionally in genetic engineering. Methods that are well known to those skilled in the art can be used to construct various plasmids and vectors; see, for example, the techniques described in Sambrook et al. (1989) and Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1989), (1994). Alternatively, the polynucleotides and vectors of the disclosure can be reconstituted into liposomes for delivery to target cells. A cloning vector may be used to isolate individual sequences of DNA. Relevant sequences can be transferred into expression vectors where expression of a particular polypeptide is required. Typical cloning vectors include pBluescript SK, pGEM, pUC9, pBR322 and pGBT9. Typical expression vectors include pTRE, pCAL-n-EK, pESP-l, pOP13CAT.
[0112] In specific embodiments, there is a vector that comprises a nucleic acid sequence that is a regulatory sequence operably linked to the nucleic acid sequence encoding MSP defined herein. Such regulatory sequences (control elements) are known to the artisan and may include a promoter, a splice cassette, translation initiation codon, translation and insertion site for introducing an insert into the vector. In specific embodiments, the nucleic acid molecule is operatively linked to said expression control sequences allowing expression in eukaryotic or prokaryotic cells.
[0113] It is envisaged that a vector is an expression vector comprising the nucleic acid molecule encoding MSP as defined herein. In specific aspects, the vector is a viral vector, such as a lentiviral vector or an adeno-associated viral vector. Lentiviral vectors are
40530616.1 commercially available, including from Clontech (Mountain View, CA) or GeneCopoeia (Rockville, MD), for example.
[0114] The term "regulatory sequence" refers to DNA sequences that are necessary to effect the expression of coding sequences to which they are ligated. The nature of such control sequences differs depending upon the host organism. In prokaryotes, control sequences generally include promoters, ribosomal binding sites, and terminators. In eukaryotes generally control sequences include promoters, terminators and, in some instances, enhancers, transactivators or transcription factors. The term "control sequence" is intended to include, at a minimum, all components the presence of which are necessary for expression, and may also include additional advantageous components.
[0115] The term "operably linked" refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" 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. In case the control sequence is a promoter, it is obvious for a skilled person that double- stranded nucleic acid is preferably used.
[0116] Thus, the recited vector is an expression vector, in certain embodiments. An "expression vector" is a construct that can be used to transform a selected host and provides for expression of a coding sequence in the selected host. Expression vectors can for instance be cloning vectors, binary vectors or integrating vectors. Expression comprises transcription of the nucleic acid molecule preferably into a translatable mRNA. Regulatory elements ensuring expression in prokaryotes and/or eukaryotic cells are well known to those skilled in the art. In the case of eukaryotic cells they comprise normally promoters ensuring initiation of transcription and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript. Possible regulatory elements permitting expression in prokaryotic host cells comprise, e.g., the PL, lac, trp or tac promoter in E. coli, and examples of regulatory elements permitting expression in eukaryotic host cells are the AOX1 or GAL1 promoter in yeast or the CMV-, SV40-, RSV- promoter (Rous sarcoma virus), CMV-enhancer, SV40-enhancer or a globin intron in mammalian and other animal cells.
40530616.1 [0117] Beside elements that are responsible for the initiation of transcription such regulatory elements may also comprise transcription termination signals, such as the SV40-poly- A site or the tk-poly-A site, downstream of the polynucleotide. Furthermore, depending on the expression system used leader sequences capable of directing the polypeptide to a cellular compartment or secreting it into the medium may be added to the coding sequence of the recited nucleic acid sequence and are well known in the art. The leader sequence(s) is (are) assembled in appropriate phase with translation, initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein, or a portion thereof, into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product; see supra. In this context, suitable expression vectors are known in the art such as Okayama-Berg cDNA expression vector pcDVl (Pharmacia), pEF-Neo, pCDM8, pRc/CMV, pcDNAl, pcDNA3 (Invitrogen), pEF-DHFR and pEF-ADA, (Raum et al. Cancer Immunol Immunother (2001) 50(3), 141-150) or pSPORTl (GIBCO BRL).
[0118] In some embodiments, the expression control sequences are eukaryotic promoter systems in vectors capable of transforming of transfecting eukaryotic host cells, but control sequences for prokaryotic hosts may also be used. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and as desired, the collection and purification of the polypeptide of the disclosure may follow. In particular embodiments, one or more encodable sequences are regulated by expression control sequences that are responsive to hypoxic environments.
[0119] Additional regulatory elements may include transcriptional as well as translational enhancers. Advantageously, the above-described vectors of the disclosure comprises a selectable and/or scorable marker. Selectable marker genes useful for the selection of transformed cells are well known to those skilled in the art and comprise, for example, antimetabolite resistance as the basis of selection for dhfr, which confers resistance to
methotrexate (Reiss, Plant Physiol. (Life-Sci. Adv.) 13 (1994), 143-149); npt, which confers
40530616.1 resistance to the aminoglycosides neomycin, kanamycin and paromycin (Herrera-Estrella, EMBO J. 2 (1983), 987-995) and hygro, which confers resistance to hygromycin (Marsh, Gene 32 (1984), 481-485). Additional selectable genes have been described, namely trpB, which allows cells to utilize indole in place of tryptophan; hisD, which allows cells to utilize histinol in place of histidine (Hartman, Proc. Natl. Acad. Sci. USA 85 (1988), 8047); mannose-6-phosphate isomerase which allows cells to utilize mannose (WO 94/20627) and ODC (ornithine
decarboxylase) which confers resistance to the ornithine decarboxylase inhibitor, 2- (difluoromethyl)-DL-ornithine, DFMO (McConlogue, 1987, In: Current Communications in Molecular Biology, Cold Spring Harbor Laboratory ed.) or deaminase from Aspergillus terreus which confers resistance to Blasticidin S (Tamura, Biosci. Biotechnol. Biochem. 59 (1995), 2336-2338).
[0120] Useful scorable markers are also known to those skilled in the art and are commercially available. Advantageously, said marker is a gene encoding luciferase (Giacomin, PI. Sci. 116 (1996), 59-72; Scikantha, J. Bact. 178 (1996), 121), green fluorescent protein
(Gerdes, FEBS Lett. 389 (1996), 44-47) or β-glucuronidase (Jefferson, EMBO J. 6 (1987), 3901- 3907). This embodiment is particularly useful for simple and rapid screening of cells, tissues and organisms containing a recited vector.
[0121] As described above, the recited nucleic acid molecule can be used in a cell, alone, or as part of a vector to express the encoded polypeptide in cells. The nucleic acid molecules or vectors containing the DNA sequence(s) encoding any one of the CD 138- specific CAR constructs is introduced into the cells that in turn produce the polypeptide of interest. The recited nucleic acid molecules and vectors may be designed for direct introduction or for introduction via liposomes, or viral vectors (e.g., adenoviral, retroviral) into a cell.
[0122] In accordance with the above, the present disclosure relates to methods to derive vectors, particularly plasmids, cosmids, viruses and bacteriophages used conventionally in genetic engineering that comprise a nucleic acid molecule encoding the polypeptide sequence of MSP defined herein. Preferably, said vector is an expression vector and/or a gene transfer or targeting vector. Expression vectors derived from viruses such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus, may be used for delivery of the
40530616.1 recited polynucleotides or vector into targeted cell populations. Methods that are well known to those skilled in the art can be used to construct recombinant vectors; see, for example, the techniques described in Sambrook et al. (loc cit), Ausubel (1989, loc cit.) or other standard text books. Alternatively, the recited nucleic acid molecules and vectors can be reconstituted into liposomes for delivery to target cells. The vectors containing the nucleic acid molecules of the disclosure can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or electroporation may be used for other cellular hosts; see Sambrook, supra.
V. Pharmaceutical Compositions
[0123] In some embodiments, there are methods for treating ALS comprising providing to an ALS patient a pharmaceutical composition comprising a pharmaceutically effective amount of MSP. Pharmaceutical compositions in accordance with certain embodiments of the present disclosure comprise an effective amount of MSP or additional active ingredient dissolved or dispersed in a pharmaceutically acceptable carrier.
[0124] As used herein, the term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio. The term "pharmaceutically acceptable carrier," means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent.
[0125] The pharmaceutical composition can be introduced to a subject by any method known to those of ordinary skill in the art. Examples may include, but not be limited to administration intravenously, intradermally, intrathecally, intraarterially, intraperitoneally, intramuscularly, subcutaneously; orally, intrarectally, mucosally (intranasal, intravaginal, etc.), topically (i.e., transdermally), locally, via inhalation (e.g., aerosol inhalation), via injection, via infusion, via continuous infusion, via localized perfusion bathing target cells directly, via a
40530616.1 catheter, via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference).
[0126] The pharmaceutical composition of the disclosure may be formulated into a composition in a free base, neutral or salt form. Pharmaceutically acceptable salts, include the acid addition salts, e.g., those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
[0127] In embodiments where the composition is in a liquid form, a carrier can be a solvent or dispersion medium comprising but not limited to, water, ethanol, polyol (e.g., glycerol, propylene glycol, liquid polyethylene glycol, etc.), lipids (e.g. , triglycerides, vegetable oils, liposomes) and combinations thereof. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin; by the maintenance of the required particle size by dispersion in carriers such as, for example liquid polyol or lipids; by the use of surfactants such as, for example hydroxypropylcellulose; or combinations thereof such methods. It may be preferable to include isotonic agents, such as, for example, sugars, sodium chloride or combinations thereof.
[0128] The actual dosage amount of a composition in accordance with certain embodiments of the present disclosure administered to an animal patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event,
40530616.1 determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
[0129] Methods may involve administering to the patient or subject at least or at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more doses of a therapeutic composition.
VI. Kits
[0130] Any of the compositions described herein may be comprised in a kit having one or more components housed in suitable container means. In specific embodiments, the kit comprises an effective amount of a MSP protein or MSP fragment or derivative. The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there are more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present disclosure also will typically include a means for containing the component(s) and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
[0131] When the components of the kit are provided in one and/or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. In some cases, the container means may itself be a syringe, pipette, and/or other such like apparatus, from which the formulation may be applied to a desired area of the body, injected into an animal, and/or even applied to and/or mixed with the other components of the kit. However, the components of the kit may be provided as dried powder(s). When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
40530616.1 [0132] Irrespective of the number and/or type of containers, the kits of the disclosure may also comprise, and/or be packaged with, an instrument for assisting with the injection/administration and/or placement of the ultimate composition within the body of an animal. Such an instrument may be a syringe, pipette, forceps, and/or any such medically approved delivery vehicle, for example.
[0133] In certain aspects, the kit comprises one or more additional agents for treatment of ALS or a symptom thereof, such as riluzole, drugs that reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, painkillers, depression treatment, treatment for sleep disturbances, and/or constipation medication, for example.
EXAMPLES
[0134] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLE 1
MSP PROTEIN AND ITS RECEPTORS IN THE THERAPY OF AMYOTROPHIC LATERAL
SCLEROSIS
[0135] Vesicle-associated membrane proteins (VAMP) are a family or proteins that mediate vesicular transport processes. VapB (Vesicle-associated membrane protein- associated protein B) is associated with the endoplasmic reticulum (ER) where it regulates protein folding and transport of ceramide (FIGS. 2-3). MSP is the N-terminal 125 amino acid domain of VapB. In the absence of VapB protein, the ER accumulates protein aggregates. The presence of the aggregates elicits a cellular stress response, the Unfolded Protein Response (UPR) in the ER. Similar protein aggregates and an UPR are also found in both sporadic and familial cases of ALS,
40530616.1 as well as in most mouse ALS models. The presence of ER aggregates and/or an UPR cause a loss or mislocalization of the VapB protein in ALS patients and mouse models for ALS. These defects lead to a decrease in the secretion of MSP, which circulates in the blood. MSP binds to multiple growth cone guidance receptors that are present on both neurons and muscles, including the EphrinA4, Dlar, and Robo receptors.
[0136] MSP binding to Ephrin A4 leads to unclustering of glutamate receptors. Conversely, loss of MSP causes a hyperclustering of glutamate receptors and an increase in Ca2+ influx, leading to neuronal excitotoxicity. Binding of MSP to Dlar and Robo is required to retain proper mitochondrial morphology, localization, and function in muscle. In the absence of MSP, muscle function is impaired. This leads to a dysfunctional motor neuron-muscle connection, resulting in a negative feed-back loop resulting from the loss of a BMP-like growth factor (GBB), which ultimately leads to neuronal loss and disease progression.
[0137] Extensive genetic, electrophysiological and biochemical studies have revealed that the Drosophila and C. elegans VapB proteins do not play an important role in synaptic transmission in vivo as originally proposed (Pennetta et al. 2002). Rather the fly, worm, and mammalian VapB homologues are type II integral membrane proteins that are mostly localized to the cytoplasm and span the ER lipid bilayer (Tsuda et al. 2008; Wyles et al. 2002). They have been implicated in a variety of processes in model organisms.
[0138] Protein aggregates are common pathological features both in familial and sporadic form of ALS. Loss of VapB in flies causes the formation of protein aggregates and an UPR in motor neurons and certain neurons in the brain (FIG. 4). This UPR is associated with a secretory defect of transmembrane proteins from ER to plasma membrane (Yang et al. 2012; Moustaqim-Barrette et al. 2013). Interestingly, an UPR has also been shown to be associated with the mutant form of VapB found in ALS8 patients (Ince et al. 2011; Papiani et al. 2012; Qiu et al. 2013; Tsuda et al. 2008) as well as numerous forms of ALS including the SODl mice models and sporadic human ALS patients (Lautenschlaeger et al. 2012). Wild type SODl has been shown to be misfolded and aggregate in sporadic ALS patients carrying the Fus mutation (R521C, Pokrishevsky et al. 2012). VapB is required for ER homeostasis.
40530616.1 [0139] VapB has significant roles in lipid metabolisms and transport. Loss of VapB causes a defect in the transport of ceramide from the ER to the Golgi (Peretti et al. 2008; Perry and Ridgway 2006). Ceramide is enriched in the ER and the spinal cord of ALS patients has been reported to have twofold excess of the normal levels of ceramide (Cutler et al. 2002).
[0140] MSP is secreted in the fly haemolymph, the worm pseudocoelom fluid, and is present in mammalian blood and functions as a hormone (Han et al. 2012; Tsuda et al. 2008). The MSP domain of VapB is evolutionarily related to a group of 28 nearly identical proteins that are abundantly expressed in C. elegans sperm (hence, the name Major Sperm Protein, MSP, Miller et al. 2001). These proteins are secreted/released from sperm by a poorly characterized mechanism (Kosinski et al. 2005) and bind to EphA4, Robo, and Lar receptors on the oocyte surface, where they regulate NMDA receptors (Miller et al. 2003; Corrigan et al. 2005). The human and fly VapB MSP domains can bind to these oocyte receptors and substitute for the functions of secreted MSPs from C. elegans sperm (Han et al. 2012; Tsuda et al. 2008). Hence, the signaling pathway is conserved between MSP and VapB proteins. Secreted MSPs from neurons in worms and flies activate growth cone guidance receptors expressed on the surface of muscles, including the EphA4, Robo, and Lar receptors (Han et al. 2012). Loss of VapB affects EphA4 signaling, causing a hyperclustering of glutamate receptors in fly muscles and
concomitant elevated Ca2+ influx (Chai et al. 2008). This in turn leads to a statistically significant increase in amplitude of electrophysiological responses in VapB loss of function mutants.
[0141] MSP/EphA4 signaling contributes to the pathology of ALS. Mammalian EphA4 can function as a receptor for MSP (Tsuda et al. 2008), and EphA4 has recently been implicated in ALS (Van Hoecke et al. 2012). EphA4 regulates actin polymerization in muscle, which is critical to the formation/stability of the neuromuscular junctions (NMJs) (Lai et al. 2001). Moreover, MSP/Lar and Robo signaling also play critical roles in the pathogenesis of the disease. Two other growth cone guidance receptors expressed on muscles, Lar and Robo, function in the control of actin polymerization and depolymerization. Moreover, the loss of these proteins or MSP severely affects mitochondrial dynamics and function in the muscles (Han et al. 2012). Significantly, similar mitochondria defects are observed in human ALS patients. The disruption in mitochondrial function and/or EphA4 receptor signaling in muscle affects the
40530616.1 retrograde signaling mechanisms from muscle to motorneurons that are critical for the proper maintenance and function of the NMJs in ALS.
[0142] SODl transgenic mice exhibit an UPR (Tobisawa et al. 2003) and a severe loss of the VapB protein (Teuling et al. 2007). This is also observed in all sporadic ALS human patients tested to date (70 out of 70), and it is the only protein that has been shown to be absent in all loci tested that cause ALS (Teuling et al. 2007; Anagnostou et al. 2010). The EphA4 receptor, which is expressed on muscle in vertebrates (Lai et al. 2001), has recently been implicated in ALS (Van Hoecke et al. 2012), and EphA4 binds MSP (Tsuda et al. 2008). Mutations in Profilin, which is required for actin polymerization, also cause ALS (Wu et al. 2012), in agreement with data showing that actin disruption in muscles affects mitochondrial dynamics. SODl transgenic mice and patients with SODl mutations exhibit a progressive loss of mitochondrial function at their NMJs and eventually exhibit defects in Ca2+ homeostasis (Zhou et al. 2010). These defects are associated with spontaneous Ca2+ spikes and muscle twitching, a feature observed in ALS patients but typically attributed to other causes. Loss of MSP leads to an increase in Ca2+ influx (via hyperclustering of glutamate receptors), alongside a concomitant decrease in Ca2+ clearing due to defective mitochondria. Together these data show that MSP is involved in proper maintenance and function of the neuromuscular junction. When compromised, the cumulative effect leads to a withdrawal of the NMJs during the final stages of the disease, ultimately leading to denervation of motor neurons that is at the core of the pathology of ALS. By binding to its cognate muscle receptors (EphA4, Dlar, and Robo), MSP promotes proper Ca2+ metabolism, mitochondrial dynamics, and mitochondrial function, preventing a premature negative feed-back loop to the presynaptic terminal. Therefore, MSP is used as a therapeutic agent in sporadic as well as familial ALS patients.
Exemplary Methods
[0143] I. Generation of Baculovirus expressing His-MSP: The human MSP domain (1st -124th amino acids of human VAPb) containing N-terminal His tag (14 amino acids) was PCR amplified by using the primers of SEQ ID NO. 1 and SEQ ID NO. 2. The PCR product was cloned into pFastBac vector [Invitrogen cat. no. 10359-016]. The pFstBac-His-MSP construct was transformed into DHlOBac E. coli. The correct recombinant bacmid clones containing His-
40530616.1 MSP were selected by PCR analysis using Ml 3 primers. The bacmid containing His-MSP was transfected into Sf9 cells to generate baculovirus expressing His-MSP protein.
[0144] II. Infection of Sf9 Cells: 5xl07 of Sf9 cells per T75 flask were seeded one hour before baculovirus infection. The cells were infected by adding ΙΟΟμΙ (MOI:~2) of baculovirus per T75 flask of Sf9 cells and incubated at 27°C for three days.
[0145] III. Protein Harvest and Purification: Three days after infection, the cells were pelleted and stored at -80°C for purification. Lysis Buffer (150 mM NaCl, 50 mM phosphate buffer pH 7.4, 10% glycerol, Roche complete protease inhibitors mix without EDTA [cat. no. 11873580001] and 1% NP40) was added to the cell pellet, which stood on ice for 30 min with occasional vortexing. The lysate from step 2 was centrifuged at 12,000 g for 30 min at 4°C. The supernatant was then collected. The supernatant was mixed with 2 ml pre- washed Ni- NTA Beads [QIAGEN; cat. no. 30210] and then rotated for 1.5 hours at 4 °C. The lysate and bead mixture from step 4 was poured into Poly-Prep Column [Bio-Rad; cat. no. 731-1550EDU] to separate the lysate and beads by gravity at 4°C (*kept the flow-through for the determination of binding efficiency). The beads were washed three times with 20ml of Wash Buffer (15 OmM NaCl, 50 mM phosphate buffer pH7.4, and 20mM imidazole) at 4 °C. The His-MSP protein was eluted with 3 ml of Elution Buffer (150mM NaCl, phosphate buffer pH 7.4, and 500 mM imidazole) at 4 °C. The eluted His-MSP was filtered at 4°C using an Amicon Ultra-30K device [Millipore; cat. no. UFC903024] to remove proteins greater than 30 KDa in size. The His-MSP was dialyzed against 1L of 1XPBS overnight twice at 4 °C. The protein concentration was measured by Bradford Assay [Bio-Rad; cat. no. 500-0006]. The protein purity was checked by 15% SDS-PAGE and coomassie blue staining.
[0146] IV. Measurement of protein stability: To measure protein stability, dialyzed His-MSP was incubated alone or with indicated amount of BSA (Sigma cat. no. A7030) at 37 °C. After incubation at 37°C for the indicated time points, the proteins were resolved by 15% SDS- PAGE and visualized by coomassie blue staining.
Table 1: Exemplary sequences
40530616.1 Sequence SEQ ID NO.
VapB makveqvlsl epqhelkfrg pftdvvttnl klgnptdrnv 1
cfkvkttapr rycvrpnsgi idagasinvs vmlqpfdydp nekskhkfmv qsmfaptdts dmeavwkeak pedlmdsklr cvfelpaend kphdveinki isttasktet pivskslsss lddtevkkvm eeckrlqgev qrlreenkqf keedglrmrk tvqsnspisa laptgkeegl strllalvvl ffivgviigk ial
MSP domain makveqvlsl epqhelkfrg pftdvvttnl klgnptdrnv 2
cfkvkttapr rycvrpnsgi idagasinvs vmlqpfdydp nekskhkfmv qsmfaptdts dmeavwkeak pedlmdsklr cvfe hMSP-F His Notl GCGGCCGCCA AAATGTCTGG TTCTCATCAT CATCATCATC 3
ATGGTAGCAG CGGCATGGCG AAGGTGGAGC AGGTC
hMSP-R Xhol CTCGAGCTAT TCAAACACAC ATCTAAGTTT TG 4
[0147] Infusion of a very low level of MSP (10-30 ng/ml) rescues the defects associated with SODl transgenic mice. At day 40, SODl (G93A) transgenic mice were surgically implanted with osmotic pump that delivered approximately 100 ng of MSP in PBS daily (saline only containing pumps serve as controls). MSP is a very stable protein at 37 °C and the pumps were reloaded every two weeks. 5/5 of the mice injected with PBS are dead whereas 3/5 of the mice injected with MSP in PBS are alive (FIGS. 6A-6B) and are still able to perform well on the rotarod assay with statistically significant differences (FIG. 7). The SODl mice injected with MSP live about 40 days longer than those injected with control saline, an extension of their lifespan by more than 25%.
EXAMPLE 2
VAPB/ALS8 MSP LIGANDS REGULATE STRIATED MUSCLE ENERGY METABOLISM
CRITICAL FOR ADULT SURVIVAL IN CAENORHABDITIS ELEGANS
[0148] ALS is a lethal neurodegenerative disease characterized by the combined degeneration of lower and upper motor neurons (Kiernan et ah, 2011). Most ALS cases occur sporadically, although a subset of cases is inherited. These familial cases are caused by mutations in multiple genes, including in the Vapb (VAMP/synaptobrevin-associated protein B) gene.
40530616.1 Mutations in Vapb lead to ALS8 that manifests as ALS or late-onset SMA, a motor neuron disease restricted to lower motor neurons (Nishimura et al., 2004; Chen et al., 2010; Kabashi et al., 2013). While Vapb mutations are rare, reduced VAPB mRNA or protein levels have been reported in sporadic ALS patients, a mSODl ALS mouse model, and ALS8 patient motor neurons derived from induced pluripotent stem cells (Anagnostou et al., 2010; Teuling et al., 2007;
Mitne-Neto et al., 2011). Hence, VAPB deficiency might have a function relevant in non-ALS8 patients.
[0149] VAPB, and its paralog VAPA, are broadly expressed type II membrane proteins that are widely conserved. These VAPs have been implicated in regulating lipid transport and homeostasis, endoplasmic reticulum (ER) dynamics, and membrane trafficking (Loewen and Levine, 2005; Amarilio et al, 2005; Lev et al, 2008; Peretti et al, 2008; De Vos et al, 2012). In addition to these cell autonomous functions, the VAP vMSP is cleaved from the transmembrane domain in the cytoplasm and secreted in a cell-type specific fashion (Tsuda et al, 2008; Han et al., 2010; Han et al., 2012). Secreted vMSPs antagonize Eph receptor signaling through a direct interaction with the extracellular domain (Tsuda et al., 2008). More recently, the inventors have shown in C. elegans and Drosophila that neurons secrete vMSPs to regulate mitochondrial localization and function in striated muscle (Han et al., 2012). vMSPs interact with muscle SAX- 3 Roundabout and CLR-1 Lar-like protein tyrosine phosphatase receptors to down-regulate CLR- 1 signaling. VAP loss causes uncontrolled CLR-1 Lar-like receptor activation in body wall muscle. CLR-1 stimulates actin filament assembly in the muscle belly that requires the actin- related protein 2/3 (Arp2/3) complex. These ectopic actin filaments displace mitochondria from I- bands, cause aberrant fission and fusion balance, and impair respiratory chain activity. Hence, neuronally- secreted vMSP promotes muscle mitochondrial localization and function, perhaps in an effort to modulate energy homeostasis.
[0150] vMSP signaling to muscle mitochondria might be relevant for the energy balance in ALS8 disease. Out of five ALS8 patients studied, five had increased cholesterol levels, four had reduced HDL, three had elevated triacylglycerol levels, and one was diabetic (Marques et al., 2006). More generally, ALS is associated with a spectrum of abnormalities in energy metabolism, including mitochondrial defects in neurons and skeletal muscle, insulin resistance,
40530616.1 dyslipidemia, and hypermetabolism (Dupuis et al, 2011). These metabolic abnormalities are positively correlated with survival. For instance, increased prediagnostic body fat is associated with decreased risk of ALS mortality (Gallo et ah, 2013) and in some patient populations, higher LDL/HDL ratios correlate with increased survival time (Dupuis et ah, 2008; Dorst et ah, 2011). However, the cause(s) of the metabolic defects and their relationship to each other are not well understood.
[0151] In embodiments herein, the inventors show in C. elegans that loss of the VAP homolog VPR-1 causes triacylglycerol (TAG) accumulation in striated body wall muscle. Mosaic analysis and tissue-specific expression studies demonstrate that VPR-1 acts in neurons, not muscle to regulate muscle fat levels. Restoring muscle mitochondrial localization and function in vpr-1 mutants decreases muscle fat content. The fat metabolism alterations are part of a compensatory response mediated by the DAF-16/FoxO transcription factor. FoxO promotes muscle fat accumulation, maintains ATP levels during aging, and extends lifespan without influencing muscle mitochondrial morphology, localization, or function. Finally, the inventors provide evidence that skeletal muscle metabolism is abnormal in Vapb mutant mice. The results indicate in the model that disrupting vMSP signaling triggers a compensatory response to muscle mitochondrial dysfunction involving FoxO transcripton factors. vpr-l/vap loss increases fat levels in adult body wall muscle and intestine
[0152] In studies of vpr-1 mutant hermaphrodites, the inventors considered that body wall muscles often contain large lipid-like droplets not observed in wild-type controls (FIG. 8A). These lipid-like droplets were visible by differential interference contrast (DIC) and transmission electron microscopy (TEM). TEM of vpr-1 (tml411) null mutant muscle shows an expanded muscle belly filled with mitochondria, as previously reported (Han et ah, 2012), and large droplets (FIG. 8B). Large droplets were not observed in TEM of wild- type muscle, vpr-1 mutants also have darker intestines than those of wildtype hermaphrodites. Dark colored intestines are seen in mutant worms with high TAG content (Watts, 2009).
[0153] To directly test whether these droplets contained fat, the inventors fed vpr-1 mutant worms E. coli incubated with Bodipy-conjugated fatty acids (Bodipy-FAs). These
40530616.1 fluorescent compounds can be used to directly visualize fat stores in live tissue (Kubagawa et al., 2006; Klapper et al., 2011). In wild-type hermaphrodite controls, dietary Bodipy-FAs were observed primarily in the intestine with a few small droplets present in muscle. In contrast, muscles of vpr-1 (tml411) null mutants contained numerous large Bodipy-FA-stained droplets (FIG. 8C). These fluorescent droplets perfectly overlapped with those observed in muscle by DIC microscopy (FIG. 8D). Bodipy-FAs are continuously transported from the diet, to the worm' s intestinal cells, and then to the muscle, where they are tightly packed in membrane -bound vesicles. Elevated fluorescence is observed in vpr-1 mutant intestine, but autofluorescent granules in this tissue makes it difficult to make definitive conclusions regarding fat content (Brooks et al., 2009). To circumvent this issue, Sudan Black B was used, which darkly stains neutral TAGs in fixed opaque worms. Sudan Black staining showed increased fat in the intestine and muscle of vpr-l(tml411 ) mutants compared to wild- type controls (FIG. 17). Finally, the inventors performed mass spectrometry of lipid extracts to determine the lipid composition of wild-type and vpr-1 mutant adult hermaphrodites. Lipids were analyzed by electrospray ionization tandem mass spectrometry spectrometry (ESTMS/MS). ESI-MS/MS analysis of the extracts detected a robust increase in TAGs in vpr-1 mutant extracts, but not in the membrane phospholipids phosphatidylethanolamine and phosphatidylcholine (FIG. 8E). These data indicate that loss of vpr-1 causes TAG accumulation in muscle and intestine of adult hermaphrodite worms.
Increased ER stress does not cause the muscle fat defect in vpr-l/vap mutants
[0154] VAP homologs have been implicated in ER stress pathways (Tsuda et al., 2008; Moumen et al., 2011; Gkogkas et al., 2008), which can modulate lipid metabolism and homeostasis (Basserti and Austin, 2012). Furthermore, mitochondrial dysfunction is sometimes associated with ER stress. The inventors considered the possibility that increased ER stress might cause the high muscle fat levels in vpr-1 mutants. Three lines of evidence argue against this possibility. First, an integrated hsp-4/BiPp::gfp ER stress reporter (Urano et al., 2002) did not show elevated stress levels in vpr-1 mutants (FIG. 18A). Second, vpr-1 mutants are not more sensitive than wild type to tunicamycin treatment, which induces ER stress (FIG. 18B). Third, RNAi of xbp-1, an ER stress-responsive transcription factor, in vpr-1 mutants had no effect on muscle fat levels in 3-day old adults (18+3.6 fat droplets/1000 μιη for vpr-l(tml411 ) [n=12]
40530616.1 versus 17.3+3.6 fat droplets/1000 μηι2 for vpr-l(tml411 ) xbp-l(RNAi) [n=10]; P = 0.28). These data indicate that increased ER stress does not cause the fat metabolism defect in vpr-1 mutants. vpr-l/vap acts cell nonautonomously to regulate fat accumulation
[0155] vpr-1 is ubiquitously expressed and its homologs have been implicated in regulating lipid dynamics via a cell autonomous mechanism (Lev et al., 2008; Wyles et al., 2002; Jansen et al., 2011; Peretti et al., 2008). To determine in which cell type(s) VPR-1 functions to regulate muscle fat, the inventors first used genetic mosaic analysis. Transgenic vpr-1 (tml411 ) mutant hermaphrodites were generated containing the vpr-1 genomic locus and the lineage marker sur-5::GFP expressed from an extrachromosomal array (Yochem and Herman, 2003). In C. elegans, extrachromosomal arrays are spontaneously lost at low frequency during cell division, thereby generating mosaic worms. When these events occur early in development, mosaic worms can be generated with losses in neurons, body wall muscles, intestinal cells, and the germ line.
[0156] Expressing the vpr-1 genomic locus in vpr-1 (tml411 ) null worms rescued the fat metabolism defect in muscle (FIG. 9), as well as the muscle mitochondrial defects, sterility, slow growth, and other phenotypes. Body wall muscles are generated from multiple cell lineages, including the EMS lineage. Transgene array loss in the EMS lineage generates mosaic worms that have a subset of muscles lacking vpr-1 expression. These muscle cells exhibited low fat levels, identical to muscle cells that express vpr-1 (FIG. 9). Therefore, VPR-1 is not required in body wall muscle. Mosaic worms lacking vpr-1 in the E lineage, which generates the intestine, also did not exhibit elevated muscle fat droplets, indicating that vpr-1 is not required in the intestine. In contrast to muscle and intestine loss, vpr-1 loss in the AB lineage, which generates the neurons, did cause increased fat droplets in muscles (FIG. 9). Unexpectedly, it was found that vpr-1 loss in the germ cell lineage causes muscle fat accumulation (FIG. 9). These results indicate that VPR- 1 acts cell nonautonomously in neurons and germ cells (or their differentiation products) to modulate fat levels in muscle.
[0157] vpr-1 null mutants are sterile, due to a failure of germ cells to differentiate into sperm and oocytes. Sperm secrete signaling molecules, such as MSPs that may influence fat metabolism (Han et al., 2010). To test whether sperm affects fat levels, the inventors mated
40530616.1 sterile 1-day-old adult vpr-l(tml411 ) hermaphrodites to wild-type males. Supplying sperm to the reproductive tract reduces muscle fat levels in vpr-l(tml411 ) mutants, as visualized with Bodipy- FAs (FIG. 19). Hence, the spermatogenesis defects in vpr- 7 mutants contribute to the muscle fat levels through a non-autonomous mechanism.
[0158] Genetic mosaics assess the effect of vpr-1 loss from cells within an otherwise vpr- background. To test whether VPR-1 expression is sufficient in neurons, the inventors expressed VPR-1 under the control of tissue-specific promoters in vpr-1 null mutants. Consistent with genetic mosaic analysis, VPR-1 expression using the myo-3 muscle-specific promoter or the ges-1 intestine- specific promoter did not influence muscle fat levels. In contrast, overexpressing vpr-1 cDNA with the unc-119 pan-neuronal promoter completely rescued the muscle fat levels in approximately 30-40% of transgenic mutant worms (FIG. 10A). Sudan Black B staining showed that fat levels were reduced in the intestine and muscle (FIG. 10B). The incomplete rescue appears to be due to the germ line defects in the transgenic mutants {i.e. lack of sperm) and missing vpr-1 introns or 3'UTR in the transgene. For instance, driving neuronal expression of the vpr-1 genomic locus instead of the cDNA rescued several mutant phenotypes with increased efficiency. These results indicate that VPR-1 acts cell non-autonomously in neurons to regulate muscle fat levels. vMSP signaling to mitochondria regulates muscle fat levels
[0159] The VAPB P56S mutation acts as a dominant negative by inhibiting secretion of the wild-type and mutant vMSPs (Tsuda et ah, 2008; Han et ah, 2012). To test whether neuronal vMSP secretion affects muscle fat levels, the inventors generated transgenic worms expressing P56S VPR-1 under the unc-119 neuronal promoter. P56S VPR-1
overexpression in wild-type worms causes increased muscle lipid droplets in most worms (FIG. 10A), indicating that vMSP secretion from neurons influences muscle fat accumulation. The inventors previously showed that impaired vMSP signaling causes ectopic Arp2/3- dependent actin filaments in muscles. A reduction of arx-2, which encodes Arp2, rescues the muscle mitochondrial defects, but not the sterility in vpr-l(tml411) mutants(Han et ah, 2012). To test whether vMSP signaling controls muscle fat metabolism, the inventors inactivated arx-2 in vpr- 1 null mutants and examined fat levels, arx-2 RNAi restored mitochondria to I-bands, as previously
40530616.1 reported (Han et al., 2012), and reduced muscle fat droplets in vpr-l(tml411 ) mutants when compared to vpr-1 (tml411 ) controls (FIG. 11; 18+3.6 fat droplets/1000 μιη2 for vpr- l(tml411 ) [n=12] versus 3.1+2 fat droplets/1000 μηι2 for vpr-l(tml411 ) arx-2(RNAi) [n=6]; P < 0.001). Identical results were observed using TEM (Han et al., 2012). Thus, in embodiments, impaired vMSP signaling from neurons to muscle mitochondria causes elevated fat levels in the intestine and body wall muscle.
DAF-16/FoxO is required for fat accumulation in vpr-l/vap mutants
[0160] The elevated TAGs in vpr-1 mutants and continuous accumulation of dietary Bodipy-FAs in muscle suggested that fat metabolism and transport pathways were altered.
Reduced energy production triggers enhanced activity of the DAF-16/FoxO transcription factor, which controls expression of genes involved in fat synthesis, fat transport, β-oxidation, and stress resistance (Oh et al., 2006; Halaschek- Wiener et al., 2005; Murphy, 2006; Murphy et al., 2003; McElwee et al., 2003). The inventors considered that the muscle mitochondrial defects may trigger elevated FoxO activity. To investigate if DAF-16 affects fat metabolism in vpr-1 mutants, they generated vpr-l(tml411) daf-16(mu86) double mutants. Muscles of daf-16(mu86) null mutants contain few Bodipy-FA-stained droplets, similar to muscles of wild-type controls.
However, muscle fat levels in the double mutants were also low, and strongly reduced when compared to those in vpr- l(tml411 ) mutants alone (FIG. 12A). daf-16 loss did not affect food intake, assessed by measuring pharyngeal pumping rates (FIG. 12B; P > 0.05), muscle mitochondria (see below), or sterility of vpr-l(tml411) mutants. In embodiments, the elevated fat levels in vpr-1 null mutants require DAF-16/FoxO activity.
[0161] The inventors next examined DAF-16/FoxO transcriptional activity using an integrated transgenic line that expresses GFP under the sod-3 promoter (sod-3p::GFP), a direct DAF-16 target (Oh et al., 2006; Henderson et al., 2006). When worms were cultured under normal growth conditions, about 40-50% of 1-day-old adult vpr-l(tml411) transgenic worms showed increased GFP expression relative to control transgenic animals (FIG. 12C). By day three of adulthood, most vpr-l(tml411 ) mutants show broad GFP expression throughout the body, including the intestine, neurons, vulva muscles, and body wall muscles. The elevated GFP expression is due to DAF-16 because GFP expression is suppressed in transgenic vpr-l(tml411)
40530616.1 daf-16(mu86) double mutants (FIG. 12C). These data indicate that vpr-1 loss causes elevated DAF-16 activity in muscles and other cell types.
[0162] To investigate the mechanism(s) by which VPR-1 controls DAF-16/FoxO, the inventors analyzed DAF-16 subcellular localization in vpr-l(tml411) mutants. An integrated and rescuing transgenic line was used that expresses DAF-16::GFP under its endogenous promoter. DAF-16::GFP translocates from cytoplasm to nucleus upon reduction in insulin signaling, although other mechanisms exist that regulate nuclear DAF-16 activity independent of translocation [39,40]. Under normal growth conditions at 20°C, DAF-16::GFP in vpr-1 mutant and control transgenic strains was distributed throughout the cytoplasm and nucleus with no significant difference between the two strains (FIGS. 13A and 13B). Hence, VPR-1 may regulate DAF-16 independent of insulin signaling. DAF-16::GFP translocation to the nucleus is observed in stressed mutant and wild-type worms, suggesting that the insulin pathway is functional (FIGS. 13A and 13B). However, vpr-1 mutants appear more sensitive to higher temperatures that require increased metabolic activity. These data support the model that VAP regulates FoxO largely through an insulin-independent mechanism.
Impaired vMSP signaling to mitochondria causes elevated DAF-16/FoxO activity
[0163] The data thus far support the model that impaired vMSP signaling to muscle mitochondria triggers elevated DAF-16 activity. Restoring muscle mitochondrial localization in vpr-1 mutants through ARX-2/Arp2 inactivation should therefore, attenuate DAF-16 activity. To assess DAF-16 transcriptional activity, the inventors used the integrated sod-3p::GFP transgenic reporter, arx-2 RNAi in vpr-l(tml411) mutants causes a strong reduction in sod-3p::GFP expression in the intestine, body wall muscle, and other cells (FIG. 13C). arx-2 RNAi in wild- type worms has little effect on GFP expression. These data support the idea that impaired vMSP signaling to muscle mitochondria increases DAF-16 activity.
[0164] To determine whether DAF-16 affects muscle mitochondria in vpr-1 mutants, the inventors first evaluated mitochondria using a transgene expressing mitochondrial matrix targeted GFP (mito::GFP). As previously documented (Han et ah, 2012), wild-type muscles contain linear mitochondrial tubules positioned along I-bands. In contrast, vpr-1 (tml411)
40530616.1 mutants contain disorganized and interconnected mitochondrial networks in the muscle belly (FIG. 14A). Loss of daf-16 in vpr-l(tml411 ) mutants did not affect muscle mitochondrial morphology or localization (FIG. 14A). Next, the inventors examined mitochondrial functional status using MitoTracker CMXRos, which accumulates in the mitochondrial matrix depending on membrane potential, and oxygen consumption of whole worms. DAF-16 loss did not affect the reduced Mitotracker CMXRos accumulation (FIG. 14B) or the low oxygen consumption rates of vpr-1 mutants (FIGS. 14C and 14D). In embodiments, DAF-16 does not influence the muscle mitochondrial defects in vpr-1 mutants and likely acts downstream of mitochondria.
DAF-16/FoxO increases ATP levels and extends lifespan of vpr-l/vap mutants
[0165] As the intestine and epidermis are fat storage sites in C. elegans, the inventors considered that the increase in muscle fat is an attempt to provide fuel for energy production. Previous studies showed that 1-day-old adult vpr-l(tml411) mutants have reduced ATP levels when compared to controls (Han et ah, 2012). However, the ATP levels in vpr-1 mutants did not decrease over the next two days, as observed in the wild type (FIG. 15A). 3-day- old adult vpr-1 (tml411) mutants had higher ATP levels than wild- type controls at the same age (FIG. 15A). Similar ATP dynamics have been observed in aging worms with mutations in the daf-2 insulin receptor or clk-1, a mitochondrial protein involved in ubiquinone biosynthesis (Houthoofd et ah, 2005; Braeckman et ah, 2002). Hence, DAF-16 may help maintain ATP levels in these aging worms. To test whether DAF-16 affects the energy balance of vpr-1 mutants, the inventors measured ATP levels in single and double mutant extracts, daf-16 loss did not influence ATP levels in 1-day-old adult vpr-l(tml411) mutants (FIG. 15A). However, daf-16 is required for the high ATP concentration in 3-day old mutant adults (FIG. 15A; P <0.001). ATP levels in daf-16 mutants are similar to wild-type controls, as previously shown (Houthoofd et ah, 2005; Braeckman et ah, 2002). These data indicate that DAF-16/FoxO helps vpr-1 mutants maintain ATP levels during aging.
[0166] Based on the abnormalities in energy metabolism, the inventors tested whether DAF-16 influences lifespan in vpr-1 mutants. Similar to other worm mutants with mild or tissue-specific reduction in mitochondrial function, vpr-l(tml411 ) mutants have slightly extended adult lifespan compared to wild-type worms (FIG. 15B; mean adult lifespan + S.D. of
40530616.1 12.9+4.4 days [n=154] for vpr-l(tml411 ) versus 10.5+2.1 days [n=159] for wild type, P < 0.001). daf-16 loss in vpr-l(tml411 ) mutants causes a strong reduction in lifespan relative to vpr-1 mutants and wild-type controls (FIG. 15B; 6.9+2.5 days for vpr-l(tml411) daf-16(mu86)
[n=250]; P < 0.001). The lifespan of daf-16 single mutants was similar to wild type, as previously shown (Murphy et ah, 2003; Lin et ah, 2201). These data indicate that DAF-16/FoxO activity extends survival of vpr-1 mutants.
Vapb knockout mice exhibit signs of abnormal skeletal muscle energy metabolism
[0167] The data thus far indicate that VPR-1 loss causes profound defects in muscle energy metabolism. The inventors considered that the regulatory function of vMSPs on energy metabolism was conserved in mammals, and studied energy metabolism of Vapb -/- mice
(Kabashi et ah, 2013). In basal conditions, Vapb -/- mice do not exhibit overt defects in energy metabolism. In particular, body weight and glycemia appear normal with age. However, an energy metabolism defect of Vapb deficient mice might be unmasked by modifying insulin supply through feeding and fasting paradigms. In worms and mice, fasting reduces insulin signaling and increases FoxO activity, resulting in altered metabolic gene expression. The inventors used Vapb -/- mice of 2-6 months of age to avoid any confounding effect of the motor dysfunction observed at 18 months(Kabashi et ah, 2013). Mice were either fasted for 24 hours (fasted group) or fasted for 16 hours and re-fed for 8 hours to synchronize meals (fed group). In +/+ mice, fasting decreased the TAG levels in the gastrocnemius (GA) muscle (FIG. 16A; P < 0.05). In contrast, TAG levels remained unchanged upon fasting in Vapb -/- GA and tibialis anterior muscles (FIG. 16A). In liver, TAG levels were unchanged upon fasting and feeding in either +/+ or -/- mice (FIG. 16A). Thus, Vapb ablation increases the resistance of muscle lipid stores to fasting induced mobilization.
[0168] The inventors next looked at mRNA levels of metabolic genes by quantitative RT-PCR. In liver, Vapb ablation potentiated induction of the direct FoxOl target gene phosphoenolpyruvate carboxykinase (PEPCK) in response to fasting, but had no effect on fasting induction of other FoxOl targets such as glucose 6-phosphatase (G6Pase) and pyruvate
40530616.1 dehydrogenase kinase (PDK4) (FIG. 16B). FoxOl and Fox03 mRNA and proteins were similar in +/+ and -/- livers, and FoxOl up-regulation by fasting appeared normal in -/- liver (FIG. 16B).
[0169] In +/+ TA muscle, feeding decreased expression of PEPCK, G6Pase, and lipoprotein lipase (LPL), and increased expression of the lipogenic transcription factor SREBPlc (FIG. 16C). This regulation was lost in Vapb -/- muscles, as feeding did not modify expression of these four genes. Vapb genotype did not affect levels of PDK4 mRNA. FoxOl and Fox03 expression was down-regulated upon feeding in control TA muscles, but Fox03 regulation was lost in -/- muscles (FIG. 16C). The expression of muscle Fox03 targets LC3 and Atroginl was up-regulated in fed -/- mice, while another Fox03 target, ATG12, was unchanged. These results indicate that muscles of Vapb -/-mice are partially insensitive to fasting/feeding alterations in lipid mobilization and FoxO target gene expression. Hence, VAPB mutant worms and mice appear to have muscle energy metabolism alterations, at least in part involving FoxO targets.
Significance of Certain Embodiments
[0170] Results from Drosophila and C. elegans support the model that VAP MSP domains are secreted neurogenic factors that promote muscle oxidative metabolism (Han et ah, 2012). In C. elegans, neurons cleave the vMSP and secrete it into the surrounding environment. Secreted vMSPs signal via SAX-3 Roundabout and CLR-1 Lar-like receptors expressed in muscle, down-regulating Lar signaling to the Arp2/3 complex. This signaling pathway restricts actin filament formation to I-bands of the myofilaments, thereby localizing mitochondria to I- bands and promoting mitochondrial function (Tsuda et ah, 2008; Han et ah, 2012). Here the inventors show that impaired vMSP signaling to muscle mitochondria triggers altered DAF- 16/FoxO transcription factor activity. FoxO increases TAG synthesis and transport to muscle, helps maintain ATP levels during aging, and extends lifespan. In some embodiments, reduced vMSP signaling puts animals in an energy deficit, which triggers an altered metabolic response involving FoxO. Evidence for this model and implications for ALS are discussed below.
The connection between VAPB and FoxO
[0171] The inventors show that vpr-l/vap loss triggers elevated DAF-16/FoxO activity in the presence of food (and insulin), resulting in muscle fat accumulation. Genetic
40530616.1 mosaic, cell-type specific expression, and genetic suppression experiments point to impaired vMSP signaling as the causal event. For example, inactivating the Arp2/3 complex in vpr-1 mutants largely suppresses DAF-16-dependent fat accumulation and SOD-3 expression. This result indicates that the cytoskeletal or mitochondrial abnormalities cause elevated DAF-16 activity in these mutants. To date, the inventors have not detected mitochondrial morphology defects in vpr-1 mutant neurons or hypodermal cells. Thus, the most likely scenario is that muscle mitochondrial dysfunction triggers elevated FoxO activity. How might this occur? The simplest model is that vpr-l/vap mutants go into energy deficit as they age. Mitochondrial dysfunction and energy deficit are thought to increase FoxO activity (Dillin et ah, 2002; Lee et ah, 2003; Billing et ah, 2011; Greer et ah, 2009). An increase in FoxO nuclear translocation is not observed in vpr- 1 mutants, at least under standard conditions. As reduced insulin levels induce FoxO
translocation, the regulation of FoxO by VPR-1 appears to be independent of insulin secretion. In mammals, FoxO transcription factors are critical regulators of energy metabolism, particularly under fasting conditions. The inventors show that Vapb ablation in mice renders muscle lipid stores resistant to fasting, a situation analogous to lipid accumulation in vpr-1 mutant worm muscles. Indeed, muscle fat persists for long time periods in starved vpr-1 mutant worms.
Dysregulated lipid stores in mutant mice is associated with alterations in muscle gene expression consistent with abnormal FoxOl and Fox03 activity (Cheng and White, 2011). For instance, FoxOl target gene mRNAs for PEPCK and G6Pase are clearly up-regulated in muscle of young Vapb -/- mice in the fed state {i.e. in the presence of insulin that decreases FoxOl activity).
Similar results are observed for Fox03 target genes LC3 and Atrogin-1. These data indicate that FoxO 1/3 are less sensitive to insulin inhibition in Vapb -/- mice.
[0172] Not all FoxO target genes studied were sensitive to Vapb ablation. For instance, VAPB does not appear to influence PDK4 and ATG12 mRNAs. Additionally, some of the mRNAs studied showed uncoupling from circulating insulin levels, consistent with an insensitivity of FoxOl to insulin. SREBPlc mRNA, which is negatively regulated by FoxOl (Kamei et ah, 2008), was increased by feeding in +/+ mice, but not in -/- mice. A similar, albeit mirror situation was observed for LPL, a gene positively regulated by FoxOl (Kamei et ah, 2003). Hence, FoxOl/3 might participate in the abnormal lipid mobilization in Vapb -/- mice, but other mechanisms are likely at work to avoid the major consequences of chronic muscle FoxO
40530616.1 activation, such as muscle atrophy (Kamei et al., 2004). In summary, the findings show that VAPB is involved in modulating mouse muscle energy metabolism upon fasting and refeeding, possibly via altered FoxO activity. Whether this occurs through a cellautonomous or, like in C. elegans and Drosophila, a cell nonautonomous mechanism remains to be determined.
FoxO is protective in vap mutants
[0173] A key finding in worms is that DAF-16/FoxO activity prolongs the adult lifespan of vpr-1 mutants from 6.9+2.5 to 12.9+4.4 days. This lifespan increase may be due to metabolic alterations that compensate for mitochondrial dysfunction. Consistent with this idea, FoxO extends the lifespan of C. elegans with reduced mitochondrial function (Dillin et al., 2002; Lee et al., 2003; Rea et al., 2007). The FoxO-dependent fat accumulation in vpr-1 mutant muscle may reflect an effort to increase energy production. The inventors show that DAF-16 helps vpr-1 mutants maintain ATP levels in 3-day old adults. Among the numerous DAF-16 metabolic genes are those involved in fat synthesis and transport, β-oxidation, the glyoxylate cycle, and gluconeogenesis (Murphy, 2006). However, additional DAF-16 targets may also be involved, such as stress resistance enzymes (Murphy, 2006; Murphy et al., 2003; Honda and Honda, 1999). vpr-1 mutants are more resistant than the wild type to reactive oxygen species and ER stress. Based on identified DAF-16 targets and vpr-1 mutant phenotypes, DAF-16 might increase energy substrate availability in muscle, stimulate anaerobic metabolism, increase oxidative metabolism in non-muscle cells, or decrease ATP consumption. Further studies are necessary to distinguish among these possibilities, as well as other models.
Implications for ALS
[0174] Metabolic alterations in ALS patients and mouse models are hypothesized to compensate for mitochondrial dysfunction, particularly in skeletal muscle (Dupuis et al., 2011; Dupuis et al., 2008; Curgnola et al., 2010; Zhou et al., 2010). Differentially expressed gene networks involved in oxidative metabolism and the cytoskeleton, including up-regulated FoxOl and FoxO3 mRNAs have been found in ALS patient skeletal muscles (Bernardini et al., 2013; Leger et al., 2006). The studies of VAPB in worms, flies, and mice are consistent with impaired vMSP signaling to muscle causing some of these alterations. Importantly, vpr-1 loss in worms,
40530616.1 vapb depletion in zebrafish, or Vapb loss in mice does not cause motor neuron degeneration (Kabashi et ah, 2013; Han et ah, 2012), providing strong evidence that mitochondrial and metabolic defects are not secondary consequences of neurodegeneration. These data contrast with a recent Drosophila study suggesting that VAPB loss causes neurodegeneration via increased phosphoinositides (Forrest et ah, 2013). In humans, metabolic alterations caused by VAPB loss may not be sufficient to induce motor neuron degeneration, although they could strongly predispose to ALS. Redundancy could be an important consideration in the different models. The worm genome encodes a single vap homolog, but many genes with MSP domains. Vertebrate genomes typically encode VAPA and VAPB, which are approximately 60% identical. Vap mutant flies have the most severe developmental defects and the fewest MSP genes in the genome.
[0175] In summary, the results indicate that striated muscle mitochondrial dysfunction alters FoxO activity, which in turn affects energy metabolism and promotes survival. In some embodiments, reduced vMSP signaling causes some of the mitochondrial and metabolic alterations in ALS patients. In certain embodiments, vMSPs protect against ALS via effects on skeletal muscle energy metabolism.
A. MATERIAL AND METHODS
C. elegans genetics, strains, and RNA-mediated interference
[0176] C. elegans Bristol N2 is the wild-type strain. Worms were grown on NGM plates with NA22 bacteria as the food source (Brenner, 1974). Strain construction and marker scoring were done as previously described (Han et ah, 2012; Miller et ah, 2013). The strains and genetic markers used or generated were as follows: CF1553 muIs84[pAD76(5,o<i-J.- :GFP)], CF1038 daf-16(mu86) I, vpr- l(tml411 )/ hT2[bli-4(e937) let-?(g782) qls48] LTII, SJ4005 zcls4[hsp-4::GFP], TJ356 zls356[daf-16p::daf-16::GFP; rol-6] IV, and XM1004 vpr-l(tml411) daf-16(mu86)l KT2[bli-4(e937) \et-l(q782) qls48] I;III. RNAi was performed using the feeding method starting at the LI stage, as previously described (Han et ah, 2012). arx-2 and xbp-1 RNAi clones are from the genome- wide library (Kamath and Ahringer, 2003). Each clone was sequenced for confirmation.
40530616.1 Transgenics
[0177] To generate transgenic C. elegans, the marker plasmids pRF4 [rol-6] (60 ng/μΐ) or myo-3p::mito::GFP (60 ng/μΐ) were mixed with myo-3p::vpr-l (60 ng/μΐ), ges-lp::vpr- 1(60 ng/μΐ), unc-119p::vpr-l (60 ng/μΐ), or unc-119p::vpr-l P56S (60 ng/μΐ) and microinjected into the gonads of young adult hermaphrodites. Injected worms were incubated for 24 hours, transferred to new NGM plates, and screened for transgenic progeny. Transgenic lines were selected based on the roller phenotype or GFP expression. Multiple independent transgenic lines were generated for all strains. To conduct genetic mosaic analysis, 10 ng/μΐ WRM06B28 fosmid DNA containing the vpr-1 genomic locus was mixed with 10 ng/μΐ pTG96 (sur-5p::GFP) plasmid and microinjected into the gonads of vpr-l(tml411 )ftiT2 hermaphrodites. Transgenic lines were selected based on GFP expression. Transgenic lines were maintained as vpr-l(tml411 ) homozygotes, as the fosmid rescued the sterility, mitochondria, fat metabolism, slow growth, and embryonic defects. For lineage scoring, approximately 15,000 worms were screened. Transgene loss in the AB lineage was scored by GFP loss in head and tail neurons, the nerve cords, and the excretory gland. Transgene loss in the PI lineage was scored by GFP loss in the intestine, muscle, somatic gonad, and hypl 1. The P2 lineage was scored by GFP loss in numerous body wall muscle cells and hypl 1, the P3 lineage was scored by GFP loss in body wall muscle, and the P4 lineage was inferred by a sterile phenotype without GFP loss. Transgene loss in the EMS lineage was scored by GFP loss in the intestine and somatic gonad, while loss in the E lineage was score by exclusive GFP loss in the intestine.
Transmission electron microscopy
[0178] TEM was performed as previously described (Han et ah, 2012). Care was taken to ensure that fixation occurred rapidly and cross sections were orthogonal to muscle myofilaments.
Bodipy-FA and Sudan Black B staining
[0179] For the Bodipy-FA experiments, a 5 mM Bodipy-FA (Molecular probe, U.S. A) stock solution was prepared in DMSO and kept at -20°C. A 200 μΜ working solution diluted in distilled water was dropped onto seeded plates and allowed to dry. L4 stage worms
40530616.1 were placed on the plates and incubated in the dark for 24 hours at 20°C. Bodipy- FAs can get trapped in intestinal gut granules that are not present in muscle. Sudan Black B staining was conducted as described in previous studies (Han et ah, 2012). Briefly, synchronized 1 -day-old adult worms were collected into microfuge tubes containing M9 solution. Worms were washed five times, incubated for 40 minutes at 20°C to remove intestinal bacteria, and fixed in 1% paraformaldehyde. The fixed worms were washed three times in cold M9 solution and dehydrated through a 25%, 50%, and 70% ethanol series. Sudan Black B solution was added to the worms and incubated for 1 hour. To remove excess stain, worms were washed five times with 70% ethanol. To normalize for staining variability among experiments, wild type and vpr-l(tml411 ) mutants were processed in the same tube and identified based on gonad morphology.
Lipid analysis by ESI-MS/MS
[0180] For the lipid analysis by ESTMS/MS, lipids from equal masses of wild type and vpr-l(tml411 ) mutant adults were extracted by chloroform-methanol following a modified Bligh/Dyer extraction (Bligh and Dyer, 1959). A mixture of internal standards including T17: l TAG was added to the chloroform-methanol phase before extraction. The extracted samples were concentrated to dryness under a nitrogen stream, reconstituted with methanokchloroform (1: 1 v/v) and transferred to HPLC auto samplers. Lipids were analyzed by ESI-MS/MS using an API 4000 (Applied Biosystems/MDS Sciex, Concord, Ontario, Canada) triple quadrupole mass spectrometer. Extracted lipid samples (5 ml) were infused into the mass spectrometer with a solvent mixture of chloroform-methanol (1:2, v/v) containing 0.1% formic acid using a Shimadzu Prominence HPLC with a refrigerated auto sampler (Shimadzu Scientific Instruments, Inc.
Columbia, MD). Lipids were analyzed in positive ion mode using an API 4000 (Applied
Biosystems/MDS Sciex, Concord, Ontario, Canada) triple quadruple mass spectrometer. Samples (5 μΐ) were directly infused into the electrospray source using a Shimadzu Prominence HPLC with a refrigerated auto sampler (Shimadzu Scientific Instruments, Inc. Columbia, MD). Neutral loss (NL) scanning (228, 254, 256, 268, 278, 280, 284, and 304) of naturally occurring aliphatic chains (i.e. building block of TAG molecular species) were utilized to determine the identities of each molecular species. NL scanning of 141 was used for profiling phosphatidylethanolamine. The following analysis parameters were used: ion spray voltage 5000 V, de-clustering potential
40530616.1 40 V, temperature 300°C (for TAG), collision energy 35 V, and collisionally activated dissociation 5.
Mitochondrial staining
[0181] To assess mitochondrial transmembrane potential, worms were stained using the MitoTracker CMXRos dye (Molecular Probes, U.S.A), as previously described (Han et ah, 2012).. This lipophilic cationic fluorescent dye accumulates in mitochondria in a membrane potentialdependent manner (Gilmore and Wilson, 1999). L4 larval stage worms were placed on dried plates containing a 100 μΜ MitoTracker CMXRos dye solution (dropped on bacteria). After 24 hours incubation in the dark, worms were transferred to a new NGM plate and incubated in the dark for 20 minutes to remove intestinal background. Worms were mounted on dried 2% agarose pads without anesthetic. Wild-type and vpr-l(tml411 ) mutant hermaphrodites were cultured on the same plates.
ATP concentration measurement
[0182] ATP concentration was measured as described previously, with slight modification (Han et ah, 2012). Briefly, 150 worms were individually picked and placed into tubes containing M9 buffer, washed four times, and incubated at 20°C for 40 minutes to remove intestinal bacteria. These worms were then washed four times with TE solution (100 mm Tris-Cl, pH 7.6, 4 mm EDTA) and placed into microfuge tubes containing 300 μΐ TE solution. Worm extracts were prepared by a series of cycles including freezing, thawing, and sonicating. These extracts were boiled for 10 minutes to release ATP and block ATPase activity. Carcasses and insoluble material were pelleted in a microcentrifuge at 20,000 x g for 10 minutes. The soluble extracts were diluted in a 1: 10 ratio using TE solution. ATP concentration in 60 μΐ of diluted extracts was measured using the ENLITEN ATP Assay System (Promega, U.S.A), according to the manufacturer's instructions. A luminometer (Berthold, Germany) was used for quantification. Protein concentration was determined using the BCA protein assay (Pierce, U.S.A). ATP measurements were repeated at least three times for each strain.
Oxygen consumption
40530616.1 [0183] Oxygen consumption rates were measured as previously described using the oxygraph system (Hansatech, UK) with minor modifications (Han et ah, 2012). Worms were cultured at 20°C and synchronized to the 1 -day-old adult stage. For each test, 1000 worms were individually picked and placed into a glass tube with 1 ml M9 buffer at 20°C. Collected worms were incubated for 40 min at 20°C to remove intestinal bacteria, carefully washed five times, and placed into 1ml M9 buffer. The worm solution was loaded into the chamber equipped with a SI Clark type polarographic oxygen electrode disc maintained at 20°C. Oxygen concentration was measured for 10 minutes. For normalization, worms were carefully collected from the chamber and protein content was measured using the BCA test kit (Pierce, U.S.A.). Rates were normalized to either total protein content or number of worms. At least three independent measurements were performed per strain.
Feeding Rate and Lifespan Assays
[0184] To measure feeding rates, worms were cultured at 20°C and 1 -day-old adult worms were placed on new NGM plates. Feeding behavior was recorded using a Zeiss Lumar stereomicro scope with AxioCam MRM digital camera. Measurements were conducted during a 30 second period at room temperature (22°C). The rhythmic contractions of the pharyngeal bulb were counted. For each strain, over 20 worms were counted. To determine lifespan of worms, L4 larval stage worms were placed on new NGM plates seeded with NA22 bacteria and cultured at 20°C. The L4 stage was used because a small percentage of vpr-1 mutants die during L1-L4 stages and vpr-1 mutants develop slowly. Worms were monitored every day and transferred to flesh NGM plates. Death was scored by failure to respond to touching with a platinum wire. Wild-type worms fed NA22 bacteria have slightly shorter lifespan than worms fed OP50 bacteria.
Mouse experiments
[0185] The mouse experiments were carried out and ethically reviewed in compliance with INSERM and French animal welfare laws, guidelines, and policies. Vapb -/- mice were used and genotyped as described(Kabashi et ah, 2013). Mice (8-10 per group) were either fasted for 24 hours from 5PM (fasted group), or fasted from 5PM to 9AM and refed until sacrifice at 5PM. Liver and muscle GA and tibialis anterior (TA) tissues were collected, and
40530616.1 rapidly frozen in liquid nitrogen for subsequent analyses of gene expression and TAG levels. The tissues were stored at -80°C until the time of analysis.
[0186] For RT-qPCR, frozen liver and muscle tissues were placed into tubes containing 5 mm stainless steel beads (Qiagen, Courtaboeuf, France) and 1 ml of Trizol reagent (Invitrogen, Paisley, UK) and homogenized using a TissueLyser (Qiagen). RNA was prepared from tissue homogenates following Trizol manufacturer's instructions. RNA reverse transcription and SYBR Green real-time PCR assays were performed using the Bio-Rad (Biorad, Marnes la Coquette, France) iCycler kits and protocols. PCR conditions were 3 min at 94°C, followed by 40 cycles of 45 s at 94°C and 10 s at 60°C. Exemplary primers are shown in Table 2.
[0187] Table 2: Exemplary PCT Primers for Respective Genes
Figure imgf000059_0001
40530616.1
Figure imgf000060_0001
[0188] For western blotting, liver and TA muscle were incubated in Lysis buffer containing complete protease and phosphatase inhibitor cocktails. Protein concentration was measured using BCA Protein Assay. Equal amount of protein (50 μg) were separated by SDS- PAGE 10% and blotted onto nitrocellulose membrane. Membranes were saturated with 10% milk and then incubated with the primary antibodies FoxOl (Proteintech™; 18592-1-AP), Fox03a (Cell signaling; #2497), VAPB [4] and Histone H3 (Cell signaling; #9715), all diluted (1: 1000) followed by anti-rabbit secondary antibody, diluted 1:5000.
[0189] For TAG analysis, tissue powder was homogenized in lysis buffer (250 mM Sucrose solution, 1 mM EDTA, 2% SDT, 1 mM DTT, 10 mM Tris HC1 pH 7.4) containing protease inhibitors (Sigma P8340) and phosphatase inhibitors (Sigma 8345), centrifuged at 12000 x rpm for 15 minutes at room temperature. TAG concentration was determined in duplicate for each sample in 5 μΐ of supernatant, using the enzymatic method of analysis (Randox Triglyceride Colorimetric Assay Kit, Randox Laboratories Limited, UK) as described by the manufacturer. Lipid values were normalized to protein concentration.
EXAMPLE 3
THE AMYOTROPHIC LATERAL SCLEROSIS 8 PROTEIN, VAP, IS REQUIRED FOR ER
PROTEIN QUALITY CONTROL
Abstract
[0190] A familial form of Amyotrophic lateral sclerosis (ALS8) is caused by a point mutation (P56S) in the VAMP associated protein B (VapB). Human VapB and Drosophila Vap- 33-1 (Vap) are homologous type II transmembrane proteins that are localized to the ER.
However, the precise consequences of the defects associated with the P56S mutation in the ER and its role in the pathology of ALS are not well understood. Here the inventors show that Vap is required for ER protein quality control (ERQC). Loss of Vap in flies shows various ERQC associated defects, including protein accumulation, ER expansion, and ER stress. It is also shown
40530616.1 that wild type Vap, but not the ALS8 mutant Vap, interacts with a lipid-binding protein,
Oxysterol binding protein (Osbp). Although loss of Osbp does not show obvious phenotypes, except male sterility, restoring expression of an Osbp that does not require Vap binding suppresses the ER defects caused by loss of Vap. Hence, in embodiments of the disclosure, the ALS8 mutation impairs the interaction of Vap with Osbp, resulting in hypomorphic defects that are similar but less severe than those observed in the vap null mutant.
Introduction
[0191] Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by preferential loss of motor neurons. Approximately 90% of all ALS cases occur sporadically, whereas the remaining 10% are inherited (Kiernan et ah, 2011; Pasinelli and Brown, 2006). Although mutations in almost 20 genes have now been shown to cause ALS (Ferraiuolo et ah, 2011), their proposed functions appear quite divergent, lacking any obvious link that would hint towards a specific molecular pathway (Al-Chalabi et ah, 2012).
[0192] ALS8 is an autosomal dominant form of ALS caused by a point mutation (P56S) in the gene encoding the VapB protein (Nishimura et ah, 2004). Human vapB is evolutionarily conserved, with homologs in numerous species (Lev et ah, 2008), including Drosophila that contains Vap-33-1 or vap. Vaps contain an amino (N)-terminal domain, called the major sperm protein (MSP) domain (Nishimura et ah, 1999; Weir et ah, 1998) and a transmembrane domain that anchors the protein in the ER (Kaiser et ah, 2005; Skehel et ah, 2000; Soussan et ah, 1999). Studies with Drosophila and C. elegans Vaps have shown that Vap has non-autonomous functions. Indeed, the MSP domain of Vap is cleaved and secreted (Han et ah, 2012; Tsuda et ah, 2008). The cleaved MSP acts as a ligand for growth cone guidance receptors expressed on muscle surfaces and affects mitochondrial dynamics in the muscles.
However, Vaps most likely also have autonomous functions as they are ER associated proteins. Indeed, they have been shown to function in glucose transport (Foster et ah, 2000), neurite extension (Matsuzaki et ah, 2011) and the development of the neuromuscular junctions (Pennetta et ah, 2002). Importantly, Vaps have also been implicated in the regulation of phospholipid biosynthetic proteins (Peretti et ah, 2008). Vaps interact with proteins containing two
phenylalanines in an acidic tract (FFAT)-motif (Mikitova and Levine, 2012) , which include lipid
40530616.1 binding proteins like Oxysterol binding protein (Osbp) (Wyles and Ridgway, 2004) and ceramide transport protein (Cert) (Kawano et ah, 2006). Studies with cultured cells indicate that the Vap/Osbp interaction is required for sphingomyelin (SM) biosynthesis in response to 25- hydroxycholesterol (Lagace et ah, 1999; Peretti et ah, 2008; Perry and Ridgway, 2006). Hence, Vap seems to be required for Osbp function at ER-Golgi membrane contact sites (Peretti et ah, 2008).
[0193] The ER is the site where newly synthesized proteins are folded and modified. Protein folding in the ER is monitored by a stringent ER quality control (ERQC) system that only permits properly folded proteins to traffic to the Golgi (Araki and Nagata, 2011; Balch et ah, 2008; Braakman and Bulleid, 2011). The accumulation of misfolded proteins in the ER caused by alterations in ER homeostasis initiates ER stress that attempts to resolve the protein-folding defects (Friedlander et ah, 2000; Travers et ah, 2000). Interestingly, ER stress has been observed in human sporadic ALS patients (Atkin et ah, 2008) and in SOD1 transgenic mice (Nishitoh et ah, 2008; Saxena et ah, 2009). Overexpression of the ALS8 mutant Vap causes ER stress in flies (Tsuda et ah, 2008). However, contrary to the findings, studies with cultured cells showed that overexpression of the ALS8 mutant Vap inhibits ER stress (Gkogkas et ah, 2008; Kanekura et ah, 2006). Hence, the precise role of Vap in ER biology remains to be determined.
[0194] To determine the function of Vap, the inventors characterized the loss of function phenotype associated with the loss of Vap in null mutant flies. It is shown that Vap is required for ER protein homeostasis. Loss of Vap causes defects in ERQC, resulting in protein accumulation and ER stress. Loss of Vap also causes Osbp to be mislocalized from ER to Golgi, and restoring expression of Osbp in the ER partially suppresses the defects caused by loss of Vap. In certain embodiments of the disclosure, loss of Vap contributes to ER stress and that this stress might play a role in the pathology of ALS.
Materials and Methods
Fly transgenes and strains
[0195] To generate UAS-osbp, the PCR fragment of the Drosophila EST clone (LD31802) containing the osbp cDNA was subcloned into pUAST and pUASTattB vector
40530616.1 (Bischof et al., 2007). To generate UAS-human OsbpL8, the PCR fragment of the human EST clone (MHS 1010-98684599) containing OsbpL8 was subcloned into attB pUASTHA vector (gift from S. Yamamoto). To generate the genomic wild type vap construct, a PCR fragment was amplified with the following primers, 5 ' -GAATTCCTTGCTTGTGGACCCCGCTGGCTG-3 ' (SEQ ID NO:27) and 5 ' -GGATACAATCTCTGCTGCTTAAATGGGATTT-3 ' (SEQ ID
NO:28). The genomic ALS8 mutant vap construct was created by chimeric PCR with primers containing a P58S mutation. The genomic fragment was subcloned into the pCasper 4 vector. To create site specific transgenes (Venken et al., 2006), the attB sequence was inserted in the genomic wild type (WT) or ALS8 mutant vap/pCasper 4 constructs. The attB constructs were injected into VK31 and VK33 attP docking sites (Venken et al., 2006).
[0196] The following strains were used:
[0197] y w, vap A31 / FM7, Kr-GAIA, UAS-GFP [vap A31] was created by imprecise excision of the P-element present in y w, P{Mae-UAS.6.11J, Vap-33-lGG01069 (Bellen et al., 2004).. Southern blotting and western blotting with the Vap (Rb92) antibody (Tsuda et al., 2008) shows that [vap A31] is a null allele. The stock containing the precise excision of y w, P{Mae-UAS.6.11J,Vap-33-lGG01069 was used as a control. The osbp null mutant was created by screening for a deletion of the DNA between two piggyBac elements, PBac{WHJOsbpf00496 and PBac(RBJe04437 (Thibault et al, 2004) using the method described by Golic and Golic (Golic and Golic, 1996) . The other strains used were y w; P(w[ +mC]= UAS- mCD8: :GFP. L /LL5(Lee and Luo, 1999) and w; P(w[+mW.hs]=GawBJD42 (D42-GAL4) (Yeh et al., 1995).
Antibody generation and immunostaining
[0198] A domain of the Osbp protein (aa 60-400) was expressed using the GST- fusion protein system. A polyclonal Guinea Pig antibody (GP89) was raised against the fusion protein at Cocalico Biologicals (Reamstown, PA). This antiserum was used at 1:3000. Larvae and adult brains were fixed in 4% paraformaldehyde for 20 minutes and washed with PBS containing 0.2% Triton X-100. The following antibody dilutions were used: rabbit anti- Vap (RB92)(Tsuda et al., 2008), 1: 1000; mouse monoclonal anti- Chaoptin (Van Vactor et al., 1988), 1:200; Elav
40530616.1 (7E8A10) (O'Neill et al, 1994), 1:200; guinea pig anti-Boca(Culi and Mann, 2003), 1:500;
guinea pig anti-Bip (Ryoo et al, 2007), 1:500 (Ryoo et al, 2007) and mouse anti-Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol {Kidd, 1998 #107)(Ryoo et al, 2007) and mouse anti- Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol {Kidd, 1998 #107)and mouse anti-Ubiquitin (FK1) (BIOMOL), 1: 100, anti-Robol (Kidd et al, 1998), 1:200 and anti-N-Cadherin (Iwai et al, 1997), 1:200. Secondary antibodies conjugated to Cy3 or Alexa 488 (Jackson ImmunoResearch, Molecular Probes) were used at 1: 1000. Images were captured with a Zeiss LSM510 confocal microscope and processed with ImageJ.
Protein chemistry
[0199] For immunoblotting, proteins from adult heads were extracted with Urea buffer (8 M urea, 4% SDS, 0.125 M Tris-HCl (pH 6.8), 12 mM EDTA, 3% β-mercaptoethanol, proteinase inhibitors (Roche), and 0.002% bromophenol blue). Anti-Vap antibody RB92, 1:5000 was used for immunoblotting.
GST Pull-Down Assays
[0200] The domain N-terminal to the transmembrane domain of the WT and ALS8 Vap protein (aal-248) was expressed using the GST-fusion protein system (Tsuda et al, 2008). To express Osbp protein, UAS- Osbp and Actin -GAL4 were transfected into Schneider cells and lysates were collected with TNT buffer (1% Triton, 50mM Tris(pH8), 150mM NaCl, ImM EDTA, proteinase inhibitors, ImM PMSF). The lysates were incubated with glutathione- Sepharose-bound WT or ALS8 Vap. The resulting precipitates were analyzed by western blotting with anti-Osbp antibodies (GP89, 1/5000) and anti-GST antibody (Sigma, 1:20,000).
Yeast Two-Hybrid Assays
[0201] WT and ALS8 human Vap protein (aal-222) were subcloned into the pBD vector (Rual et al, 2005). Human OsbpL8 and Orp3 were subcloned into pAD vector (Rual et al, 2005). All interaction assays were performed by co-transfecting the two vectors encoding the hybrid proteins into the yeast strain Mav203 and plating on selective media lacking histidine. PGAL assays were performed as described previously (Rual et al, 2005).
40530616.1 Transmission Electron Microscopy
[0202] TEM was performed as previously described (Verstreken et al., 2003). Flight assay
[0203] Flight assays were performed as previously described (Fayyazuddin et al. , 2006; Rual ei al, 2005).
Giant Fiber recording
[0204] Giant Fiber recordings were performed with a protocol modified from (Tanouye and Wyman, 1980). Briefly, flies were anaesthetized on ice, transferred to a petri dish filled with soft dental wax, and the fly wings and legs were mounted in wax, ventral side down, using forceps. For stimulations and recordings from the TTM (TergoTrochanter Muscle) and DLM (Dorsolateral Muscle), five electrolytically sharpened tungsten electrodes were used: two for stimulating the giant fiber (GF), one as a reference electrode, and two for recording from the TTM and DLM respectively. To activate the GF, two sharp tungsten electrodes were inserted into each eye and voltage stimulation was applied at different frequency stimulations. GF-DLM and GF-TTM responses were measured through two electrodes implanted in the DLM and TTM. Prior to applying high frequency stimulation on the GF, low frequency stimulations at 0.5Hz were applied after placing the two recording electrodes in TTM and DLM to ensure that the electrodes are recording from the proper muscles (the latency of responses for TTM is 0.8ms and for DLM 1.2ms; Tanouye and Wyman, 1980). High frequency train stimulations of 20 pulses were delivered to the GF at 10Hz, 20Hz, 50Hz, 100Hz and 200Hz in random order. Ten times repetitive stimulations were applied for each particular frequency train, interspersed with five minutes rest between two trains of stimuli. 0.5Hz stimulations were used again after high frequency stimulation to confirm that electrodes were still in the proper muscle. Stimuli of the crossing electrodes were fixed at a duration of 0.1ms at 8-15 V of amplitudes through a stimulus isolation unit (model DS2A, Digitimer Ltd, England) and the frequency of train stimuli was controlled by AxoGraph acquisition software (AxoGraph Scientific). A microelectrode amplifier (Model 1800, A-M system) was used for all recordings. Digidata 1322A (Axon Instruments) was used for data acquisition. The probability of responses, under particular frequency of GF
40530616.1 stimulation, due to a particular stimulus (nth, n=l, 2, ..., 20), was calculated from the proportion of successful responses (out of 10 of repetitive, note: not 20; also see S10-C) for both TTM and DLM pathways. The difference of probability of responses between control and mutant (p value) for particular stimuli were calculated by a t-test (SigmaPlot 10; Systat Software, Inc.).
Results
Vap is required for ER proteostasis
[0205] The MSP domain of Vap (MSP Vap) is cleaved and functions as a secreted ligand for muscle expressed Eph, Robo, and Dlar receptors in flies and worms (Han et ah, 2012; Tsuda et ah, 2008). Vap is also localized at the ER and overexpression of the ALS8 mutant isoform causes ER stress in flies (Tsuda et ah, 2008), suggesting that Vap may play a role in ER stress as well. It was therefore examined if Vap is required for ER proteostasis.
[0206] The ER is integral to maintaining protein homeostasis (proteostasis), as protein-folding of transmembrane and secreted proteins occurs under the supervision of ERQC (Vembar and Brodsky, 2008). The ERQC is able to identify misfolded proteins, retrotranslocate the misfolded proteins and promote their degradation. ERQC overload induces ER stress, which restores proteostasis by halting protein translation, and by activating signaling pathways that lead to an increased production of molecular chaperones, which facilitate protein folding (Bernales et ah, 2006). The ERQC is important especially for membrane proteins, which are prone to aggregation due to their inherent tendency to assemble in oligomers (Hurtley and Helenius, 1989).
[0207] To determine if Vap functions in ER proteostasis, it was examined if membrane proteins accumulate in the neurons of the vap null mutant animals (Pennetta et al. , 2002). Adult brains were stained of control and vap null mutant flies with an antibody for Chaoptin, a membrane protein that is expressed in photoreceptor cells (Van Vactor et ah, 1988). In neurons of the adult cortex of control flies, cortical neurons in the brain express low levels of Chaoptin, which has not been previously reported (FIG. 20A, arrows). However, in vap mutants, Chaoptin accumulates in punctae in the cytoplasm of the cell body of these cortical neurons (FIG. 20B, B' and B", arrows). In addition, other membrane proteins such as Robo-1 (Seeger et
40530616.1 al., 1993) and N-Cadherin (Iwai et al., 1997) also accumulate in the cytoplasm of the soma of neurons of vap null mutants (FIG. 20D and F), but not in control flies (FIG. 20C and E). These accumulated proteins are surrounded by ER membranes, as shown in neurons of the adult brain that are co-stained with anti-Chaoptin and anti-Boca antibody, an ER marker (Culi and Mann, 2003) (FIG. 21A, A' and A"). Interestingly, these proteins only accumulate in a relatively small subpopulation of neurons (FIGS. 20 and 21).
[0208] To test if a specific neuronal population is affected in the vap mutants the inventors expressed a membrane anchored GFP (CD8-GFP) protein (Wong et al., 2002) driven by a broadly expressed neuronal GAL4 driver (OK307-GAL4) (Fayyazuddin et al., 2006) and stained adult cortical neurons with anti-Chaoptin and anti-GFP antibodies (FIGS. 2B, B' and B"). Although CD8-GFP is expressed in all cortical neurons (FIGS. 2B and B'), only a selective subset of neurons exhibits an accumulation of both Chaoptin and CD8-GFP proteins (FIGS.2B, B'and B", arrows), suggesting that specific subtypes of neurons are vulnerable to the defects caused by loss of vap. Furthermore, although CD8-GFP is accumulated in the cytoplasm of vap null adult flies, the inventors did not observe these accumulations in mutant larvae, indicating that protein accumulations are stage and possibly age dependent. Thus, loss of Vap causes
accumulation of membrane proteins associated with the ER in a stage-dependent and neuronal cell type specific manner, suggesting that Vap might be required for ER proteostasis.
Loss of Vap causes ER stress
[0209] Defects in ER proteostasis have been found to cause ER stress (Friedlander et al., 2000; Travers et al., 2000). To determine the ultrastructural features associated with loss of Vap, the inventors carried out transmission electron microscopy (TEM) of adult brains of wild type and mutant flies. These analyses revealed large intracellular vacuoles in a subset of neurons, often near the nuclei (FIGS. 22A-B, 100% of the brains exhibit vacuoles in a few percent of the neurons in 3 vap null mutants versus 0% in 3 brains of controls). These vacuoles are surrounded by a single membrane and contain non-homogenous materials (asterisk, FIG. 22B). In addition, the vacuoles are contiguous with the nuclear membrane (FIG. 22C, arrows) and decorated with electron-dense ribosomes (FIG. 22D, arrow heads), indicating that they correspond to a vastly expanded ER. These ER expansions are typically observed in the presence of ER stress (Schuck
40530616.1 et al, 2009), which induces ire -1 -mediated unconventional splicing of the Drosophila xbpl mRNA (Calfon et al, 2002). Upon expression of the ER stress sensor Xbpl-GFP (Kang and Ryoo, 2009; Ryoo et al, 2007) in control and vap mutant flies, GFP is significantly upregulated in a subpopulation of mutant neurons (FIG. 23B and D), but not in control flies (FIG. 23A and C). Similarly, Bip, a chaperone and ER stress indicator (Ryoo et al, 2007), is also upregulated in mutant brains relative to control flies (FIG. 23E and F), further suggesting that loss of Vap causes ER stress. In summary, the data suggest that loss of Vap causes defects in ER proteostasis resulting in accumulation of membrane proteins and ER stress in a subpopulation of neurons in the fruitfly brain.
Loss of Vap causes accumulation of ubiquitinated proteins
[0210] Continuous ER stress causes an impairment of the proteasome system and leads to accumulation of ubiquitinated proteins (Menendez-Benito et al, 2005). Moreover, ubiquitin immunoreactive inclusions in lower motor neurons represent a characteristic
pathological feature of ALS (Leigh et al., 1991). The inventors therefore assessed the presence of ubiquitinated proteins in adult brains using an ubiquitin antibody. Ubiquitinated proteins accumulate in subpopulation of cortical neurons of adult mutant neurons (FIG. 23H and H'), but not in brains of larvae and control adults (FIG. 4G and G'). Hence, the data suggest that loss of Vap leads to accumulation of ubiquitinated proteins.
VAP is required for the proper localization of OSBP
[0211] Neurons in which the ALS8 protein is overexpressed also exhibit an ER stress, similar to what is observed in the vap null mutant ((Tsuda et al, 2008) and FIG. 23). The inventors therefore examined if the ALS8 mutation causes a loss of interactions with proteins that are required for its function. The inventors performed a two hybrid screen using a human adult brain cDNA library to identify proteins that are able to bind to the WT human protein, but are unable to interact with the ALS 8 human Vap mutant protein when used as a bait (Rual et al, 2005). Among the positives there were two OSBP related proteins: ORP3 (Gregorio-King et al, 2001; Lehto et al, 2005; Lehto et al, 2008) and OSBPL6 (Lehto and Olkkonen, 2003). Both interact with wild type VapB, but are not able to interact with ALS 8 VapB as reported previously
40530616.1 (Teuling et al, 2007). The Drosophila Osbp homolog encoded by CG6708 contains a Vap binding site or FFAT-motif (Alphey et al, 1998; Ma et al, 2010). As shown in FIG. 24A,
Drosophila WT Vap, but not the ALS8 mutant version, can interact with Osbp in GST pull down assays, indicating that the interaction between Vap and Osbp is direct and evolutionarily conserved. The Osbp protein family is an evolutionarily conserved protein family whose function was initially linked to non-vesicular intracellular transport of sterols in yeast (Raychaudhuri and Prinz, 2010; Ridgway, 2010; Yan and Olkkonen, 2008). However, recent studies suggest that Osbps integrate sterol and sphingomyelin metabolism (Banerji et al, 2010; Goto et al, 2012; Yan and Olkkonen, 2008), as well as control of microtubule-dependent motility of
endosomes/lysosomes, and exocytosis (Raychaudhuri and Prinz, 2010). Studies with cultured cells indicate that the Vap/Osbp interaction is required for sphingomyelin (SM) biosynthesis in response to 25-hydroxycholesterol (Lagace et al, 1999; Peretti et al, 2008; Perry and Ridgway, 2006). Vap seems to be required for Osbp function in SM biosynthesis at ER- Golgi membrane contact sites (Peretti et al, 2008).
[0212] To determine the cellular distribution of Osbp, the inventors generated antisera against Drosophila Osbp (GP89). To confirm the specificity of this antibody (GP89), null alleles of Osbp (see Material and Methods) were created. The inventors confirmed the deletion of the osbp locus with southern blotting and the loss of Osbp protein expression with western blotting in the mutants (FIG. 24B). As shown in FIG. 24C and C, loss of Osbp in mutant clones leads to a loss of staining in GFP positive mutant clones, showing that the antibody is specific. To examine the cellular distribution of Osbp and Vap, salivary glands were co-stained with anti-Osbp and anti-Vap antibody. As shown in FIG. 24D, Osbp colocalizes with Vap in the ER and the cell membrane.
[0213] To determine whether Vap is required for the proper localization and function of Osbp the localization of Osbp was examined in vap null mutant clones. The inventors performed MARCM analysis (Lee et al, 2000) to create mutant vap clones and labeled them with the Osbp antibody. As shown in FIG. 25, loss of vap (marked by GFP) leads to an aberrant distribution of Osbp and an accumulation of Osbp in punctae (arrows in FIG. 6B'). Co-staining the vap null mutant neurons with a Golgi marker, the Msl20 kDa antibody (Stanley et al, 1997)
40530616.1 (FIG. 25 C-D), revealed that Osbp (FIG. 25D') co-localizes with Msl20 (FIG. 6D"). In the absence of vap, Osbp is mislocalized from ER to Golgi.
ALS8 mutation causes a partial loss of function of Vap
[0214] Overexpression of ALS8 mutant Vap (VapALS8 ) in flies affects the secretion of the MSP aminoterminal domain of VapALS8, causing a loss of function of Vap. Furthermore, expression of VapALS8 causes an ER stress when overexpressed in neurons.
However, overexperession of VAPALS8 also rescues the lethality associated with the loss of Vap. The data suggest that VapALS8 is a partial loss of function mutation, and that
overexpression may mask or create some of the associated cell biological defects (Tsuda et ah, 2008). To obtain the proper tissue specificity and express Vap and VapALS8 proteins at physiological levels, the inventors created transgenic flies carrying a genomic fragment of vap using P-element mediated transformation with and without a site specific attB docking site to attenuate positional effects of transgenes (Venken et ah, 2011). These constructs were
transformed using classical P-element mediated transformation or phi-C31 mediated
transformation in the VK31 and VK33 docking sites (Groth et ah, 2004); Venken et ah, 2006). Western blots confirmed that both the VapWT and VapALS8 proteins encoded by these transgenes are expressed at levels that are comparable with endogenous Vap (FIG. 26A).
[0215] The inventors first tested if the genomic vapWT can rescue the lethality associated with the vap null mutants (Avap) (FIG. 26B). Loss of vap causes pupal or pharate adult lethality with occasional adult escapers. All tested genomic vapWT transgenes (4 lines) rescue the lethality associated with loss of Vap and restore normal lifespan of the flies (FIGS. 26B and C). Eight out of eleven P-element vapALS8 transgenic lines also rescued the lethality, suggesting that VapALS8 protein retains some WT protein function (FIGS. 26B and C).
However, all VapALS8 rescued flies exhibit a severely reduced lifespan when compared to VapWT rescued flies (FIG. 26B and C, compare Avap; vapWT (F7), 52 days and Avap; vapALS8 (M6), 23 days; * P<0.001). Furthermore, an additional copy of vapWT (Avap; vapWT ' / vapALS8 ) compensates for the defects associated with vapALS8 (FIG. 26B; compare the lifespan of Advap; vapALS8 (M6), 23 days and Avap ;vapWT/vapALS8 (M6), 65days; * P<0.001), suggesting that the ALS8 mutation only causes a partial loss-of-function.
40530616.1 [0216] To avoid possible positional effects of the different transgenes, the ability was compared of vapWT and vapALS8 transgenes inserted in the same docking site (VK31 line) to rescue the vap null associated phenotypes. The inventors confirmed that vapALS8 rescued flies indeed have a severely reduced lifespan when compared to vapWT rescued flies (FIG. 26B,
Δναρ; vapALS8 (VK31 line), 14 days vs. Δναρ; vapWT (VK31 line), 67days), showing that position effects are not responsible for the observed differences in life span. To assess the physiological consequences of the incomplete rescue flight ability (FIG. 26D), brain pathology (FIG. 27A), and the electrophysiological properties of adult neurons was examined using the giant fiber responses of Avap;vapALS8 (VK31) transgenic flies (FIGS. 27B and C). Interestingly, vapALS8 rescued flies exhibit a progressive flight defect (FIG. 26D) that worsens with age.
However, an additional copy of vapWT suppresses the defects associated with vapALS8 (FIG. 26D, compare Δναρ; vapALS8 and Δναρ / vapWT; vapALS8), suggesting that the defects caused by the ALS8 mutation are due to a partial loss of function. Hence, vapALS8 causes defects in flight ability in an age dependent manner.
[0217] To assess the morphological consequences of the Δναρ flies rescued with vapALS8, histological sections were examined of adult brains that are 12 days old. As shown in FIG. 27A, there were very significant histological differences in these brains when compared to those of the proper controls. The adult brains of Δναρ; vapWT flies did not show any obvious defects (FIG. 27A). In contrast, Δναρ; vapALS8 flies exhibit numerous vacuoles in the optic lobe and central brain (FIG. 27A), but they are more frequent in the central lobe. Importantly, an additional copy of vapWT completely suppresses the defects associated with vapALS8, suggesting that the ALS8 mutation is less potent than the WT copy.
[0218] To assess neuronal function in the adults, there were recordings from the giant fiber system (GFS). The giant fiber system consists of large neurons whose input consist of visual and mechanosensory inputs (Tanouye and Wyman, 1980). Within the GFS, there are two types of motor neurons that are activated by a stimulus that induces escape behavior: the dorsal longitudinal flight motor and tergotrochanteral jump motor neurons. Their activation leads to contractions of the dorsal longitudinal muscles (DLMs) and tergotrochanteral muscles (TTMs) respectively (Pavlidis and Tanouye, 1995; Tanouye and Wyman, 1980). To stimulate the GFS
40530616.1 one impales stimulation electrodes in the eyes and records from the different muscles simultaneously (Pavlidis and Tanouye, 1995; Tanouye and Wyman, 1980). Following GF stimulation, responses were monitored of the DLM and TTM using different stimulation protocols (from 10Hz to 200Hz with fixed 20 pulses, see Methods). The DLM and TTM responses accurately reflect the activity of the motor neurons (Koenig and Ikeda, 1983; Pavlidis and Tanouye, 1995), providing a read-out of motor neuron function in the adult, allowing to assess an age dependent demise of the function of these neurons. As shown in FIG. 27B, control flies can maintain responses in TTM and DLM at the 20th pulse of a 200Hz stimulation. In contrast, one day old vap null mutants are unable to follow increasing stimulation rates, suggesting that loss of VAP causes severe functional defects in the adult motor neuron system (FIG. 27B). However, the neuronal network must be intact as the flies still respond to a 10 Hz stimulation rather efficiently at day 1 when vap is lost.
[0219] To determine the rescuing effects of VAPALS8 in adult motor neurons, the inventors also performed GF recordings of Δναρ; vapWT and Δναρ; vapALS8 (FIG. 27C).
Importantly, the vapWT transgene can fully rescue the defects in Δναρ adult motor neurons (FIG. 27C). However, vapALS8 rescues the defects only partially (day 6) and exhibits a defect that progressively worsens (Δναρ; vapALS8, 6 days vs. 12 days), suggesting that vapALS8 causes a progressive demise of the GFS. In summary, the data suggest that VapALS8 is less potent than VapWT and hence, the vapALSS mutation is a partial loss of function allele of vap. osbp genetically interacts with the ALS8 mutation
[0220] To determine if loss of OSBP shows similar defects as observed when VAP is lost, the inventors tested the phenotype associated with loss of osbp (Adosbp) (Parks et ah, 2004). Flies lacking osbp (Adosbp/Df(3R)ED622), are viable and male-sterile, in agreement with previously published observations (Ma et ah, 2010). However, in contrast to loss of vap, loss of osbp does not lead to the accumulation of ubiquitinated proteins or ER stress. These observations suggest that osbp plays a redundant role with other osbp paralogues in flies, as reported previously (Ma et ah, 2010).
40530616.1 [0221] The Osbp is severely mislocalized in vap null mutant cells (FIG. 25), and that the protein is mis-localized to the Golgi, rather than the ER. To determine if the
mislocalization contributes to defects in the ER when Vap is lost, transgenic flies were created carrying the human OsbpL8 (Yan et ah, 2008). Human OsbpL8 contains a conserved Osbp domain and an ER retention signal, but unlike Osbp and human Osbp6, it does not contain the FFAT binding motif that is required for binding to the VAP protein (FIG. 28G) (Yan et al. , 2008), suggesting that human OsbpL8 may be able to function in the ER in the absence of vap. Loss of Vap causes increased levels of Bip protein (compare FIG. 9C to FIG. 9A) and accumulation of ubiquitinated proteins (FIG. 9D compared to FIG. 9B). These defects are accompanied by the mislocalization of Osbp (FIG. 9D' compared to FIG. 9B'). To determine if restoring expression of an Osbp that does not require the presence of VAP binding alleviates the defects associated with loss of VAP, human OsbpL8 HA tagged protein was expressed in neurons of the vap null mutant. As expected, human OsbpL8 distributes diffusely in the cytoplasm of neurons in the vap null mutant, wild type flies, and heterozygous vap mutant control flies (compare FIG. 28F' to FIG. 28B' and 28H'). Hence, human OsbpL8 localization, unlike Osbp, does not depend on Vap expression. Interestingly, human OsbpL8 strongly attenuates the upregulation of Bip and the accumulation of ubiquitinated proteins caused by loss of Vap (FIGS. 26E and 26F compared to 26C and 26D), showing that restoring expression of Osbp is able to suppress the defects associated with loss of Vap. Taken together, these data suggest that loss of Vap causes the mislocalization of Osbp which in turn mediates the ERQC defects.
Significance of Certain Embodiments
[0222] Based on the findings, in embodiments of the disclosure an impairment of the normal function of Vap in the ER may contribute to the pathology of ALS8. Vap is required for the proper localization of Osbp in the ER. The interaction between Vap and Osbp function is apparently required for the implementation of the ERQC. Failure of Vap-Osbp to function causes a defect in ER proteostasis, resulting in protein accumulations in the ER and ER stress. These defects in the ER overload probably promote the Ubiquitin Proteasome System (UPS), which subsequently leads to an accumulation of ubiquitinated proteins. In the autosomal dominant form of ALS, ALS8, patients express both WT Vap and the ALS8 mutant Vap. As the ALS8 mutant
40530616.1 Vap is not able to bind to Osbp, this loss of binding seems to result in partial loss of function of Vap. This in turn may cause a gradual decrease in function of the ERQC. These defects may also lead to secondary defects that have been previously reported: namely the lack of the secretion of MSP (Tsuda et al, 2008; Han et al, 2012).
[0223] Recently defects in ER proteostasis have been implicated in the pathology of ALS. The ER constantly requires maintenance of protein homeostasis, or proteostasis (Brodsky and McCracken, 1999). To this end, the ER carries the burden of continuously modulating protein folding and degradation to avoid accumulation of misfolded proteins (Vembar and Brodsky, 2008). To deal with the challenge of protein mis-folding, eukaryotic cells have evolved an ERQC mechanism. An overloaded ERQC induces ER stress, and severe or prolonged ER stress leads to cell dysfunction and eventually cell death. Various proteins involved in ERQC have been identified, including VCP (Latterich et al, 1995), Derlin-l(Lilley and Ploegh, 2004) and
Calreticulin (Parlati et al, 1995), and a familial form of ALS is associated with mutations in VCP (Johnson et al., 2010). Moreover, mutations in SOD1 have been shown to inhibit the function of Derlin-1 (Nishitoh et al., 2008) and reduce the levels of Calreticulin and trigger ER stress (Bernard-Marissal et al., 2012). In SOD1 mice, the ER stress seems to contribute to selective motor neuron degeneration (Nishitoh et al., 2008; Saxena et al., 2009), and ER stress has also been implicated in patients with sporadic ALS (Atkin et al., 2008). Together, these observations suggest that defects in ER proteostasis might be a common pathological feature of ALS.
Interestingly, VapB levels decrease concomitantly with the disease's progression in the SOD1 mouse model (Teuling et al., 2007), and sporadic ALS patients have been reported to have decreased levels of the VapB protein (Anagnostou et al., 2010; Teuling et al., 2007), suggesting that impaired VAPB function may contribute to the pathogenesis of familial and sporadic forms of ALS. It is therefore possible that the molecular mechanism by which loss of Vap causes defects may be not restricted to ALS 8.
[0224] An important question raised by the results is how VapB functions in protein homeostasis in the ER. The ERQC is involved in identifying aberrantly misfolded proteins, retrotranslocating the misfolded proteins and processing the degraded retrotranslocated proteins (Vembar and Brodsky, 2008). These processes seem to be tightly linked. Indeed, many proteins
40530616.1 function in multiple different steps in the ERQC. However, Vap is unlikely to function in chaperone-dependent refolding, since a molecular chaperone, Bip, is upregulated in vap null mutants. Moreover, Bip overexpression fails to rescue the ER stress in the vap null mutants.
[0225] Restoring the levels of Osbp in the ER suppresses the ER defects caused by loss of Vap, suggesting that this pathway is required for the execution of the ERQC. Mammalian oxysterol binding protein (Osbp) and OSBP-related protein (Orp) constitute a large eukaryotic gene family characterized by a conserved C-terminal sterolbinding domain (FIG. 28G) (Lehto and Olkkonen, 2003). This domain organization suggests that a primary function of Osbp/Orp is to transfer cholesterol or oxysterols between target membranes, and/or transduce sterol-dependent signals at these points of contact (Banerji et ah, 2010; Ngo and Ridgway, 2009; Suchanek et ah, 2007). Differential localization of Osbp between organelles in response to exogenous and endogenous sterol ligands suggests that Osbp transfers cholesterol and/or oxysterols between these organelles. Although the endoplasmic reticulum (ER) membrane is cholesterol poor (3-6% of total lipids) (Lange, 1991), acute cholesterol depletion in culture medium impairs the mobility of membrane proteins and thus protein secretion from the ER in cultured cells (Ridsdale et ah, 2006). Hence, the defects in ER proteostasis might be due to decreased levels of cholesterol in the ER caused by loss of Osbp/Vap function. In addition, Osbps are also coupled to the activation of ceramide transport protein (Cert) and sphingomyelin (SM) synthesis through increased activity of PI4KIIa, a cholesterol sensitive PI 4-kinase (Banerji et ah, 2010; Waugh et ah, 2006). This suggests that loss of Vap may also cause defects in ceramide transport from the ER, which may result in accumulation of ceramide and defects in ER proteostasis, consistent with the observation that ceramide accumulates in ER stress (Swanton et ah, 2007). Moreover, the mislocalization of Osbp may also affect the plasma membrane (PM), phosphatidylinositol 4-phosphate (PI(4)P) levels (Stefan et ah, 2011), the PM sterol distribution (Georgiev et ah, 2011) and polarized endocytosis (Alfaro et ah, 2011).
[0226] Previous studies have suggested that the ALS8 mutant Vap proteins cause dominant negative defects (Kanekura et ah, 2006; Han et ah, 2012). Interestingly, RNAi knockdown of VapB attenuated the ER stress associated with ALS8 overexpression (Kanekura et ah, 2006). However, these assays were performed by transient transfection of the mutant Vap
40530616.1 gene and RNAi into cultured cell lines. It is therefore likely that neurons in vivo respond differently to this acute stress (Kikuchi et ah, 2006). Indeed, flies expressing the ALS8 mutant protein at physiological levels and in the appropriate tissues show that the flies can typically live for about two weeks before their nervous system function starts to break down and death ensues, indicating that the ALS8 mutant protein retains some function and that it causes hypomorphic defects rather than gain of function defects. Moreover, these data are also consistent with the previous observations in vivo that overexpression of ALS8 causes ER defects and act as a dominant negative when overexpressed (Han et ah, 2012). In certain embodiments, then, loss of ALS8 and the ALS8 mutation cause similar phenotypes and the ALS8 mutation in patients is a partial loss of function mutation, i.e. VapB is haploinsufficient in humans. In summary, studying the normal function of the ALS8 gene has advanced the understanding of the molecular, physiological, and pathophysiological aspects of the disease, and ER stress plays a role in numerous forms of ALS.
EXAMPLE 4
MSP ACTIVATES TYROSINE KINASE RECEPTORS
[0227] In one embodiment of the disclosure, a model shows that MSP is secreted in blood and binds to growth cone guidance receptors, including ROBO and a LAR like phosphor- tyrosine phosphatase, to modulate muscle mitochondrial dynamics. It has been shown that ALS patients have reduced level of VapB and that these correlate with neuronal and muscle impairments. Therefore, it was considered that MSP serves as a hormone to control the health or disease status of neurons and muscles, in particular embodiments. To address the signaling function of MSP, human HEK293 cells were treated with purified recombinant His-tagged MSP (His-MSP), and the overall phosphor- tyro sine profile was examined through western blotting. Addition of His-MSP up-regulates tyrosine phosphorylation signals in HEK293 cells (FIG. 29, A). Protein antibody microarray data reveal an increased in protein phosphorylation of receptor tyrosine kinases, such as EGFR, FGFR and the Eph receptors (FIG. 29, B & C) as well as downstream signaling molecules, inculding AKT, ERK1/2 and Src (FIG. 29, B & C). Bioinformatics analysis of these targets predicts a signaling network regulated by MSP (FIG. 29, D), in certain embodiments of the disclosure. This is consistent with a role of MSP as a hormone.
40530616.1 [0228] To further dissect the biological function of secreted MSP, it was tested whether MSP treatment promotes growth of the cultured primarily hippocampal neurons. There was an up-regulation in the bassoon positive synapses in these primarily cultured neurons upon His-MSP treatment (FIG. 30), indicating that the biological activity of MSP regulates synapse formation and/or maintenance in neurons, in certain embodiments of the disclosure. Taken together, the data indicates that MSP modulates complex protein phosphorylation signaling networks that control synapse formation in hippocampus neurons.
[0229] Procedures for an exemplary Phosphor-tyrosine blot is as follows:
[0230] 1. The day before the assay, 90% confluent HEK293 cells were changed to serum free culture medium.
[0231] 2. After 16 hours serum starve, HEK293 cells were stimulated with lOOng/ml of His-MSP at 37 °C incubator for the indicated time points.
[0232] 3. The HEK293 cells were washed twice with ice-cold 1XPBS and then lysed in Lysis Buffer (20mM Hepes pH7.5, 150mM NaCl, 1%NP40, 50mM NaF, ImM Na3V04, 10% glycerol, protease inhibitor cocktail from Roche).
[0233] 4. After cell lysis, the total lysate was harvested by centrifuged at 14,000 rpm for 10 minutes.
[0234] 5. Total protein amount was measured by Bradford Method (Bio-RAD). [0235] 6. Equal amount of protein lysate was resolved in 10% SDS-PAGEs. [0236] 7. The total proteins were transferred to nitrocellular paper for Western bolts.
[0237] 8. After transfer, the nitrocellular paper was blocking with Blocking solution (5% Bovine Serum Albumin in IX TBST) at room temperature for 1 hour.
[0238] 9. Anti-phosphotyrosine antibody (clone 4G10; Millipore) was used as first antibody to detected phosphotyrosine proteins.
40530616.1 [0239] 10. Goat anti-mouse IgG conjugated with HRP was used to detect the signaling of the first antibody.
[0240] 11. ECL kit was used the highlight the phosphotyrosine proteins.
[0241] An exemplary Protein Phosphorylation Microarray (PathScan RTK Signaling Antibody Array Kit; Cell Signal) is as follows:
[0242] The assays were done following the procedures provided by the kit listed as below:
[0243] 1. Bring glass slides and blocking buffer to room temperature before use.
[0244] 2. Prepare IX Array Wash Buffer by diluting 20X Array Wash Buffer in deionized water. Keep at room temperature. Dilute 1 mL of 20X Array Wash Buffer with 19 mL of deionized water. Label as IX Array Wash Buffer.
[0245] 3. Prepare IX Detection Antibody Cocktail as follow: For running only 1 slide: Dilute 150 of 10X Detection Antibody Cocktail with 1350 of Array Diluent Buffer. For running 2 slides: Dilute 300 μΐ^ of 10X Detection Antibody Cocktail with 2700 μΐ^ of Array Diluent Buffer. *Keep on ice.
[0246] 4. Prepare IX HRP-linked Streptavidin as follow: For running only 1 slide: Dilute 150 of 10X HRP-linked Streptavidin with 1350 μΐ, of Array Diluent Buffer. For running 2 slides: Dilute 300 μΐ^ of 10X HRP linked Streptavidin with 2700 μΐ^ of Array Diluent Buffer. *Keep on ice.
[0247] 5. Affix the multi-well gasket to the glass slide:
[0248] a. Place the multi-well gasket face-down on the benchtop (the silicone layer should be facing up). Remove the protective plastic film.
[0249] b. Carefully place the glass slide on top of the multi-well gasket with the nitro-cellulose pads facing down while aligning the pads with the openings in the gasket. The orientation line should appear in the upper left hand corner when the slide is oriented vertically.
40530616.1 [0250] c. Insert the numbered metal clip into the groove in the gasket and rotate the clip into the locked position. Ensure that the clip is on the same side as the orientation line on the slide.
[0251] d. Slide the clip into place. The number "1" on the metal clip will now be in the same corner of the assembly as the orientation line.
[0252] e. Snap the unmarked metal clip to the other side of the assembly in the same manner and slide into place.
[0253] f. The assembled array is ready to use.
[0254] 6. Add 150 μΐ Array Blocking Buffer to each well and cover with sealing tape. Incubate for 15 minutes at room temperature on an orbital shaker.
[0255] Note: Do not allow the pads to dry out at any time during the assay.
[0256] 7. Decant Array Blocking Buffer by gently flicking out the liquid into a sink or other appropriate waste receptacle. Add 150 μΐ diluted lysate to each well and cover with sealing tape. Incubate for 2 hours at room temp (or overnight at 4°C) on an orbital shaker.
[0257] 8. Decant well contents by gently flicking out the liquid into a sink or other ap-ipropriate waste receptacle. Add 200 μΐ (IX) Array Wash Buffer to each well and incubate for 5 minutes at room temperature on an orbital shaker. Repeat three more times. Decant well contents.
[0258] 9. Add 150 μΐ (IX) Detection Antibody Cocktail to each well and cover with sealing tape. Incubate for 1 hour at room temperature on an orbital shaker.
[0259] 10. Wash 4 X 5 minutes with 200 μΐ (IX) Array Wash Buffer as in step 8.
[0260] 11. Add 150 μΐ ( IX) HRP-linked Streptavidin to each well and cover with sealing tape. Incubate for 30 minutes at room temperature on an orbital shaker.
[0261] 12. Wash 4 X 5 minutes with 200 μΐ (IX) Array Wash Buffer as in step 8.
40530616.1 [0262] 13. Remove multi-well gasket by pulling the bottom of the metal clips away from the center of the slide, then peeling the slide and gasket apart.
[0263] 14. Place the slide face up in a plastic dish (a clean pipette tip box cover works well). Wash briefly with 10 ml (IX) Array Wash Buffer.
[0264] 15. Dilute and combine LumiGLO® and Peroxide reagents immediately before use (to make 10 ml of a IX solution, combine 9 ml deionized water with 0.5 ml of 20X LumiGLO® and 0.5 ml of 20X Peroxide).
[0265] 16. Decant Array Wash Buffer and cover slide with LumiGLO®/Peroxide reagent.
[0266] 17. Transfer slide to sheet protector, ensuring that it is still covered by LumiGLO®/Peroxide reagent (add a small amount on top of the slide).
[0267] 18. Immediately capture an image of the slide on a film.
[0268] An exemplary Bioinformatics analysis of MSP signaling network is as follows:
[0269] STRING9.05 program (http://string-db.org/) was used. The proteins hits isolated from the microarray assays were input to the program. The default settings were used to generate the signaling network.
[0270] Exemplary primary hippocampal cultures were as follows:
[0271] 1 million primary hippocampal neurons were obtained from Primary Cell, LLC. Neurons were plated on PDL-coated glass covered cover slips with MEM/FGF medium as described by the manufacturer with a cell density of 1.6x10 4 cells/cm 2. 4 hours after plating, medium was replaced with pre-balanced NB27G - maintenance medium. Medium was changed every 3-4 days and replaced with pre-balanced medium. 13 day post plating, hippocampal neurons were treated with either lOOng/ml MSP or equal volume of vehicle (PBS). After 5 days of treatment, cells were washed with PBS and fixed for 15 minutes in 4% PF A/PBS at room
40530616.1 temperature. Cells were washed 3 times in lx PBS for 10 minutes each prior to blocking for 1 hour in blocking solution (10 % NGS, 0.3 % Triton-X, PBS, pH 7.4). After blocking, cultures were incubated with primary antibody (Guinea pig Bassoon 1: 1000, Synaptic Systems 141004, Chicken Map2 1:500) overnight at 4 degrees. The following day, cultures were washed 4 times in lx PBS for 5 minutes each prior to incubation with secondary antibody (anti-Guinea pig Cy5, 1: 1000 (Jackson ImmunoResearch 706-175-148), anti-Chicken Cy3, 1: 1000 (Jackson
ImmunoResearch 303-165-003) in blocking solution). After 1 hour at room temperature, cultures were washed once again 4 times 5 minutes in lx PBS and mounted on glass coverslips using Vectashield mounting medium with DAPI (Vector Labs). For each well, images of 10 separated fields of view of separate population of neurons were taken using a Leica confocal microscope under a 20 x objective, zoom factor 3x, Z-plane 8.1 μιη. The number synapses on 250 μιη dendrites were quantified for each image.
EXAMPLE 5
VAPB GENE THERAPY FOR ALS
[0272] The present example describes studies utilizing a polynucleotide encoding VAPB for therapeutic application in a mouse model for ALS. Described herein are two main outcome measures that showed improvement following viral expression of VAPB in the brains of mice of mouse model of ALS.
[0273] The mouse model for ALS utilized is the SOD1 G93A transgenic mouse purchased from the Jackson Laboratories (B6.Cg-Tg(SODl*G93A)lGur/J) and outcrossed to FVB to produce FVBB6 offspring for study. FIG. 31 illustrates an exemplary viral expression vector for injection into the SOD1 G93A transgenic mouse, wherein the construct encodes full- length VAPB and a label, such as yellow fluorescent protein (YFP), for example. In specific embodiments, MSP domain of VAPB or a functionally active fragment or derivative of MSP, are injected into the SOD1 G93A transgenic mouse. FIG. 32 illustrates an example of an injection regiment for studies in the ALS mouse model.
[0274] To measure the effects of VAPB in the ALS mouse model, RotaRod testing was used to assess outcome. In RotaRod testing, mice are placed on a rod and the rod is rotated
40530616.1 for 5 minutes, with some acceleration. Time may be recorded either when the mice fall or when they rotate twice around the rod. As an example, more than one trial per day may be given to the mice, and multiple training days may be included in a given week. In a specific embodiment of the testing, three trials a day were given for experimental and control mice, including for three training days, followed by once a week thereafter. Five-minute trials were performed, with acceleration of the rod from 5-40 rpm, with no reverse motion.
[0275] FIG. 33 shows RotaRod testing for control and SOD mice given AAV8- VAPB or left untreated. The decline in motor performance measured by rotarod testing is temporarily slowed in the mice treated with VAPB. FIG. 34 illustrates survival rate of
VAPB/SOD1 G93A transgenic mice, wherein the experimental mice had an extended life compared to control mice.
[0275] A second measure included a behavioral score, which is a crude measure of limb control used in the art. The scores utilized for measure (described by The Jackson
Laboratory; also see Vercelli et al (2008)) are as follows:
[0277] Score 5; Normal movement, Full extension of hind legs away from lateral midline when mouse is suspended by its tail;
[0278] Score 4; Collapse or partial collapse of leg extension towards lateral midline or trembling of hind legs during tail suspension;
[0279] Score 3; Toes curl under at least twice during walking of 12 inches, or any part of foot is dragging along cage bottom/table;
[0280] Score 2; Rigid paralysis or minimal joint movement, foot not being used for forward motion; and
[0281] Score 1; Mouse cannot right itself within 30 seconds from either side.
[0282] FIG. 35 shows exemplary behavioral scores in VAPB-treated SOD mice compared to untreated SOD control mice lacking VAPB overexpression. The VAPB-treated SOD mice decline more slowly at early points of disease than untreated SOD controls.
40530616.1 [0283] FIG. 36 shows that there is strong overexpression of VAPB protein in the brain of SODl/VAPB transgenic mice, and FIG. 37 demonstrates increased levels of MSP fragment in the plasma of wild-type mice treated with the same AAV8-VAPB vector.
40530616.1 REFERENCES
Al-Chalabi, A., Jones, A., Troakes, C, King, A., Al-Sarraj, S., and van den Berg, L.H. (2012). The genetics and neuropathology of amyotrophic lateral sclerosis. Acta
neuropathologica 124, 339-352.
Alfaro, G., Johansen, J., Dighe, S.A., Duamel, G., Kozminski, K.G., and Beh, C.T. (2011). The sterol-binding protein Kesl/Osh4p is a regulator of polarized exocytosis. Traffic 12, 1521-1536.
Alphey, L., Jimenez, J., and Glover, D. (1998). A Drosophila homologue of oxysterol binding protein (OSBP)— implications for the role of OSBP. Biochimica et biophysica acta 1395, 159-164.
Anagnostou G, Akbar MT, Paul P, Angelinetta C, Steiner TJ, de Belleroche J (2010) Vesicle associated membrane protein B (VAP) is decreased in ALS spinal cord. Neurobiol Aging 31:969-85.
Araki, K., and Nagata, K. (2011). Protein folding and quality control in the ER. Cold Spring Harbor perspectives in biology 3, a007526.
Atkin, J.D., Farg, M.A., Walker, A.K., McLean, C, Tomas, D., and Home, M.K. (2008). Endoplasmic reticulum stress and induction of the unfolded protein response in human sporadic amyotrophic lateral sclerosis. Neurobiology of disease 30, 400-407.
Balch, W.E., Morimoto, R.I., Dillin, A., and Kelly, J.W. (2008). Adapting proteostasis for disease intervention. Science 319, 916-919.
Banerji, S., Ngo, M., Lane, C.F., Robinson, C.A., Minogue, S., and Ridgway, N.D. (2010). Oxysterol binding protein-dependent activation of sphingomyelin synthesis in the golgi apparatus requires phosphatidylinositol 4-kinase Ilalpha. Mol Biol Cell 21, 4141-4150.
Bellen, H.J., Levis, R.W., Liao, G., He, Y., Carlson, J.W., Tsang, G., Evans-Holm, M., Hiesinger, P.R., Schulze, K.L., Rubin, G.M., et al. (2004). The BDGP gene disruption
40530616.1 project: single transposon insertions associated with 40% of Drosophila genes. Genetics 167, 761-781.
Bernales, S., Papa, F.R., and Walter, P. (2006). Intracellular signaling by the unfolded protein response. Annual review of cell and developmental biology 22, 487-508.
Bernard-Marissal, N., Moumen, A., Sunyach, C, Pellegrino, C, Dudley, K.,
Henderson, C.E., Raoul, C, and Pettmann, B. (2012). Reduced calreticulin levels link
endoplasmic reticulum stress and Fas-triggered cell death in motoneurons vulnerable to ALS. The Journal of neuroscience : the official journal of the Society for Neuroscience 32, 4901-4912.
Bischof, J., Maeda, R.K., Hediger, M., Karch, F., and Basler, K. (2007). An optimized transgenesis system for Drosophila using germ-line- specific phiC31 integrases.
Proceedings of the National Academy of Sciences of the United States of America 104, 3312- 3317.
Braakman, I., and Bulleid, N.J. (2011). Protein folding and modification in the mammalian endoplasmic reticulum. Annual review of biochemistry 80, 71-99.
Brodsky, J.L., and McCracken, A. A. (1999). ER protein quality control and proteasome-mediated protein degradation. Seminars in cell & developmental biology 10, 507- 513.
Calfon, M., Zeng, H., Urano, F., Till, J.H., Hubbard, S.R., Harding, H.P., Clark, S.G., and Ron, D. (2002). IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415, 92-96.
Corrigan C, Subramanian R, Miller MA (2005) Eph and NMDA receptors control Ca2+/calmodulin-dependent protein kinase II activation during C. elegans oocyte meiotic maturation. Development 132, 5225-37.
Culi, J., and Mann, R.S. (2003). Boca, an endoplasmic reticulum protein required for wingless signaling and trafficking of LDL receptor family members in Drosophila. Cell 112, 343- 354.
40530616.1 Cutler RG, Pedersen WA, Camandola S, Rothstein JD, Mattson MP (2002) Evidence that accumulation of ceramides and cholesterol esters mediates oxidative stress-induced death of motor neurons in amyotrophic lateral sclerosis. Ann Neurol 52, 448-57.
Fayyazuddin, A., Zaheer, M.A., Hiesinger, P.R., and Bellen, H.J. (2006). The nicotinic acetylcholine receptor Dalpha7 is required for an escape behavior in Drosophila. PLoS biology 4, e63.
Ferraiuolo, L., Kirby, J., Grierson, A. J., Sendtner, M., and Shaw, P.J. (2011).
Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nature reviews Neurology 7, 616-630.
Foster, L.J., Weir, M.L., Lim, D.Y., Liu, Z., Trimble, W.S., and Klip, A. (2000). A functional role for VAP-33 in insulin- stimulated GLUT4 traffic. Traffic 1, 512-521.
Friedlander, R., Jarosch, E., Urban, J., Volkwein, C, and Sommer, T. (2000). A regulatory link between ER-associated protein degradation and the unfolded-protein response. Nature cell biology 2, 379-384.
Georgiev, A.G., Sullivan, D.P., Kersting, M.C., Dittman, J.S., Beh, C.T., and Menon, A.K. (2011). Osh proteins regulate membrane sterol organization but are not required for sterol movement between the ER and PM. Traffic 12, 1341-1355.
Gkogkas, C, Middleton, S., Kremer, A.M., Wardrope, C, Hannah, M., Gillingwater, T.H., and Skehel, P. (2008). VAPB interacts with and modulates the activity of ATF6. Human molecular genetics 17, 1517-1526.
Golic, K.G., and Golic, M.M. (1996). Engineering the Drosophila genome: chromosome rearrangements by design. Genetics 144, 1693-1711.
Goto, A., Liu, X., Robinson, C.A., and Ridgway, N.D. (2012). Multisite phosphorylation of oxysterol-binding protein regulates sterol binding and activation of sphingomyelin synthesis. Mol Biol Cell 23, 3624-3635.
40530616.1 Gregorio-King, C.C., Collier, G.R., McMillan, J.S., Waugh, CM., McLeod, J.L., Collier, F.M., and Kirkland, M.A. (2001). ORP-3, a human oxysterol-binding protein gene differentially expressed in hematopoietic cells. Blood 98, 2279-2281.
Groth, A.C., Fish, M., Nusse, R., and Calos, M.P. (2004). Construction of transgenic Drosophila by using the site-specific integrase from phage phiC31. Genetics 166, 1775-1782.
Gurney ME, Pu H, et al. (1994) Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation. Science 264, 1772-5.
Han, S.M., Tsuda, H., Yang, Y., Vibbert, J., Cottee, P., Lee, S.J., Winek, J., Haueter, C, Bellen, H.J., and Miller, M.A. (2012). Secreted VAPB/ALS8 Major Sperm Protein Domains Modulate Mitochondrial Localization and Morphology via Growth Cone Guidance Receptors. Dev. Cell., 22:348-62.
Hiesinger, P.R., Fayyazuddin, A., Mehta, S.Q., Rosenmund, T., Schulze, K.L., Zhai, R.G., Verstreken, P., Cao, Y., Zhou, Y., Kunz, J., et al. (2005). The v-ATPase V0 subunit al is required for a late step in synaptic vesicle exocytosis in Drosophila. Cell 121, 607-620.
Hurtley, S.M., and Helenius, A. (1989). Protein oligomerization in the endoplasmic reticulum. Annual review of cell biology 5, 277-307.
Ince PG, Highley JR, Kirby J, Wharton SB, Takahashi H, Strong MJ, Shaw PJ (2011) Molecular pathology and genetic advances in amyotrophic lateral sclerosis: an emerging molecular pathway and the significance of glial pathology. Acta Neuropathol 122, 657-71.
Iwai, Y., Usui, T., Hirano, S., Steward, R., Takeichi, M., and Uemura, T. (1997). Axon patterning requires DNcadherin, a novel neuronal adhesion receptor, in the Drosophila embryonic CNS. Neuron 19, 77-89.
Johnson, J.O., Mandrioli, J., Benatar, M., Abramzon, Y., Van Deerlin, V.M., Trojanowski, J.Q., Gibbs, J.R., Brunetti, M., Gronka, S., Wuu, J., et al. (2010). Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 68, 857-864.
40530616.1 Kaiser, S.E., Brickner, J.H., Reilein, A.R., Fenn, T.D., Walter, P., and Brunger, A.T. (2005). Structural basis of FFAT motif-mediated ER targeting. Structure 13, 1035-1045.
Kanekura, K., Nishimoto, I., Aiso, S., and Matsuoka, M. (2006). Characterization of amyotrophic lateral sclerosis-linked P56S mutation of vesicle-associated membrane protein- associated protein B (VAPB/ALS8). The Journal of biological chemistry 281, 30223-30233.
Kang, M.J., and Ryoo, H.D. (2009). Suppression of retinal degeneration in Drosophila by stimulation of ER-associated degradation. Proceedings of the National Academy of Sciences of the United States of America 106, 17043-17048.
Kawano, M., Kumagai, K., Nishijima, M., and Hanada, K. (2006). Efficient trafficking of ceramide from the endoplasmic reticulum to the Golgi apparatus requires a VAMP- associated protein-interacting FFAT motif of CERT. The Journal of biological chemistry 281, 30279-30288.
Kidd, T., Brose, K., Mitchell, K.J., Fetter, R.D., Tessier-Lavigne, M., Goodman, C.S., and Tear, G. (1998). Roundabout controls axon crossing of the CNS midline and defines a novel subfamily of evolutionarily conserved guidance receptors. Cell 92, 205-215.
Kiernan, M.C., Vucic, S., Cheah, B.C., Turner, M.R., Eisen, A., Hardiman, O., Burrell, J.R., and Zoing, M.C. (2011). Amyotrophic lateral sclerosis. Lancet 377, 942-955.
Kikuchi, H., Aimer, G., Yamashita, S., Guegan, C, Nagai, M., Xu, Z., Sosunov, A.A., McKhann, G.M., 2nd, and Przedborski, S. (2006). Spinal cord endoplasmic reticulum stress associated with a microsomal accumulation of mutant superoxide dismutase-1 in an ALS model. Proceedings of the National Academy of Sciences of the United States of America 103, 6025- 6030.
Koenig, J.H., and Ikeda, K. (1983). Evidence for a presynaptic blockage of transmission in a temperaturesensitive mutant of Drosophila. Journal of neurobiology 14, 411- 419.
40530616.1 Kosinski M, McDonald K, Schwartz J, Yamamoto I, Greenstein D (2005) C. elegans sperm bud vesicles to deliver a meiotic maturation signal to distant oocytes. Development 132, 3357-69.
Lagace, T.A., Byers, D.M., Cook, H.W., and Ridgway, N.D. (1999). Chinese hamster ovary cells overexpressing the oxysterol binding protein (OSBP) display enhanced synthesis of sphingomyelin in response to 25-hydroxycholesterol. Journal of lipid research 40, 109-116.
Lai KO, Ip FC, Cheung J, Fu AK, Ip NY (2001) Expression of Eph receptors in skeletal muscle and their localization at the neuromuscular junction. Mol Cell Neurosci 17, 1034- 47.
Lange, Y. (1991). Disposition of intracellular cholesterol in human fibroblasts.
Journal of lipid research 32, 329-339.
Latterich, M., Frohlich, K.U., and Schekman, R. (1995). Membrane fusion and the cell cycle: Cdc48p participates in the fusion of ER membranes. Cell 82, 885-893.
Lautenschlaeger J, Prell T, Grosskreutz J (2012) Endoplasmic reticulum stress and the ER mitochondrial calcium cycle in amyotrophic lateral sclerosis. Amyotwph Lateral Scler 13, 166-77.
Lee, T., and Luo, L. (1999). Mosaic analysis with a repressible cell marker for studies of gene function in neuronal morphogenesis. Neuron 22, 451-461.
Lee, T., Winter, C, Marticke, S.S., Lee, A., and Luo, L. (2000). Essential roles of Drosophila RhoA in the regulation of neuroblast proliferation and dendritic but not axonal morphogenesis. Neuron 25, 307-316.
Lehto, M., and Olkkonen, V.M. (2003). The OSBP-related proteins: a novel protein family involved in vesicle transport, cellular lipid metabolism, and cell signalling. Biochimica et biophysica acta 1631, 1-11.
40530616.1 Lehto, M., Hynynen, R., Karjalainen, K., Kuismanen, E., Hyvarinen, K., and
Olkkonen, V.M. (2005). Targeting of OSBP-related protein 3 (ORP3) to endoplasmic reticulum and plasma membrane is controlled by multiple determinants. Experimental cell research 310, 445-462.
Lehto, M., Mayranpaa, M.I., Pellinen, T., Hialmo, P., Lehtonen, S., Kovanen, P.T., Groop, P.H., Ivaska, J., and Olkkonen, V.M. (2008). The R-Ras interaction partner ORP3 regulates cell adhesion. Journal of cell science 121, 695-705.
Leigh, P.N., Whitwell, H., Garofalo, O., Buller, J., Swash, M., Martin, J.E., Gallo, J.M., Weller, R.O., and Anderton, B.H. (1991). Ubiquitin-immunoreactive intraneuronal inclusions in amyotrophic lateral sclerosis. Morphology, distribution, and specificity. Brain : a journal of neurology 114 { Ft 2), 775-788.
Lev, S., Ben Halevy, D., Peretti, D., and Dahan, N. (2008). The VAP protein family: from cellular functions to motor neuron disease. Trends in cell biology 18, 282-290.
Lilley, B.N., and Ploegh, H.L. (2004). A membrane protein required for dislocation of misfolded proteins from the ER. Nature 429, 834-840.
Ma, Z., Liu, Z., and Huang, X. (2010). OSBP- and FAN-mediated sterol requirement for spermatogenesis in Drosophila. Development 137, 3775-3784.
Matsuzaki, F., Shirane, M., Matsumoto, M., and Nakayama, K.I. (2011). Protrudin serves as an adaptor molecule that connects KIF5 and its cargoes in vesicular transport during process formation. Mol Biol Cell 22, 4602-4620.
Menendez-Benito, V., Verhoef, L.G., Masucci, M.G., and Dantuma, N.P. (2005). Endoplasmic reticulum stress compromises the ubiquitin-proteasome system. Human molecular genetics 14, 2787-2799.
Mikitova, V., and Levine, T.P. (2012). Analysis of the key elements of FFAT-like motifs identifies new proteins that potentially bind VAP on the ER, including two AKAPs and FAPP2. PloS one 7, e30455.
40530616.1 Miller MA, Nguyen VQ, Lee MH, Kosinski M, Schedl T, Caprioli RM, Greenstein D (2001) A sperm cytoskeletal protein that signals oocyte meiotic maturation and ovulation. Science 291, 2144-7.
Miller MA, Ruest PJ, Kosinski M, Hanks SK, Greenstein D (2003) An Eph receptor sperm-sensing control mechanism for oocyte meiotic maturation in Caenorhabditis elegans. Genes Dev 17, 187-200.
Moustaqim-Barrette A, Tsuda H, Bellen HJ (2013) The Amyotrophic Lateral Sclerosis 8 protein, VAP, is required for ER protein quality control. Submitted.
Ngo, M., and Ridgway, N.D. (2009). Oxysterol binding protein-related Protein 9 (ORP9) is a cholesterol transfer protein that regulates Golgi structure and function. Mol Biol Cell 20, 1388-1399.
Nishimura, A.L., Mitne-Neto, M., Silva, H.C., Richieri-Costa, A., Middleton, S., Cascio, D., Kok, F., Oliveira, J.R., Gillingwater, T., Webb, J., et al. (2004). A mutation in the vesicle-trafficking protein VAPB causes late onset spinal muscular atrophy and amyotrophic lateral sclerosis. American journal of human genetics 75:822- 831.
Nishimura, Y., Hayashi, M., Inada, H., and Tanaka, T. (1999). Molecular cloning and characterization of mammalian homologues of vesicle-associated membrane protein-associated (VAMP-associated) proteins. Biochemical and biophysical research communications 254:21-26.
Nishitoh, H., Kadowaki, H., Nagai, A., Maruyama, T., Yokota, T., Fukutomi, H., Noguchi, T., Matsuzawa, A., Takeda, K., and Ichijo, H. (2008). ALS-linked mutant SOD1 induces ER stress- and ASKl-dependent motor neuron death by targeting Derlin-1. Genes & development 22, 1451-1464.
O'Neill, E.M., Rebay, I., Tjian, R., and Rubin, G.M. (1994). The activities of two Ets- related transcription factors required for Drosophila eye development are modulated by the Ras/MAPK pathway. Cell 78, 137-147.
40530616.1 Papiani G, Ruggiano A, Fossati M, Raimondi A, Bertoni G, Francolini M, Benfante R, Navone F, Borgese N (2012) Restructured endoplasmic reticulum generated by mutant amyotrophic lateral sclerosis-linked VAPB is cleared by the proteasome. J Cell Sci 125, 3601-11.
Parks, A.L., Cook, K.R., Belvin, M., Dompe, N.A., Fawcett, R., Huppert, K., Tan, L.R., Winter, C.G., Bogart, K.P., Deal, J.E., et al. (2004). Systematic generation of high- resolution deletion coverage of the Drosophila melanogaster genome. Nature genetics 36, 288- 292.
Parlati, F., Dominguez, M., Bergeron, J. J., and Thomas, D.Y. (1995). Saccharomyces cerevisiae CNE1 encodes an endoplasmic reticulum (ER) membrane protein with sequence similarity to calnexin and calreticulin and functions as a constituent of the ER quality control apparatus. The Journal of biological chemistry 270, 244-253.
Pasinelli, P., and Brown, R.H. (2006). Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nature reviews Neuroscience 7, 710-723.
Pavlidis, P., and Tanouye, M.A. (1995). Seizures and failures in the giant fiber pathway of Drosophila bangsensitive paralytic mutants. The Journal of neuroscience : the official journal of the Society for Neuroscience 15, 5810-5819.
Pennetta, G., Hiesinger, P.R., Fabian-Fine, R., Meinertzhagen, LA., and Bellen, H.J. (2002). Drosophila VAP- 33A directs bouton formation at neuromuscular junctions in a dosage- dependent manner. Neuron 35:291-306.
Peretti, D., Dahan, N., Shimoni, E., Hirschberg, K., and Lev, S. (2008). Coordinated lipid transfer between the endoplasmic reticulum and the Golgi complex requires the VAP proteins and is essential for Golgi-mediated transport. Mol Biol Cell 19:3871-3884.
Perry, R.J., and Ridgway, N.D. (2006). Oxysterol-binding protein and vesicle- associated membrane protein-associated protein are required for sterol-dependent activation of the ceramide transport protein. Mol Biol Cell 17:2604-2616.
40530616.1 Pokrishevsky E, Grad LI, Yousefi M, Wang J, Mackenzie IR, Cashman NR (2012) Aberrant localization of FUS and TDP43 is associated with misfolding of SODl in amyotrophic lateral sclerosis. PLoS One 7, e35050.
Qiu L, Qiao T, Beers M, Tan W, Wang H, Yang B, Xu Z (2013) Widespread aggregation of mutant VAPB associated with ALS does not cause motor neuron degeneration or modulate mutant SODl aggregation and toxicity in mice. Mol Neurodegener 8, 1.
Raychaudhuri, S., and Prinz, W.A. (2010). The diverse functions of oxysterol-binding proteins. Annual review of cell and developmental biology 26, 157-177.
Ridgway, N.D. (2010). Oxysterol-binding proteins. Sub-cellular biochemistry 51,
159-182.
Ridsdale, A., Denis, M., Gougeon, P.Y., Ngsee, J.K., Presley, J.F., and Zha, X.
(2006). Cholesterol is required for efficient endoplasmic reticulum- to-Golgi transport of secretory membrane proteins. Mol Biol Cell 17, 1593-1605.
Rual, J.F., Venkatesan, K., Hao, T., Hirozane-Kishikawa, T., Dricot, A., Li, N., Berriz, G.F., Gibbons, F.D., Dreze, M., Ayivi-Guedehoussou, N., et al. (2005). Towards a pro teome- scale map of the human protein-protein interaction network. Nature 437, 1173-1178.
Ryoo, H.D., Domingos, P.M., Kang, M.J., and Steller, H. (2007). Unfolded protein response in a Drosophila model for retinal degeneration. The EMBO journal 26, 242-252.
Saxena, S., Cabuy, E., and Caroni, P. (2009). A role for motoneuron subtype- selective ER stress in disease manifestations of FALS mice. Nature neuroscience 12, 627-636.
Schuck, S., Prinz, W.A., Thorn, K.S., Voss, C, and Walter, P. (2009). Membrane expansion alleviates endoplasmic reticulum stress independently of the unfolded protein response. The Journal of cell biology 187, 525-536.
40530616.1 Seeger, M., Tear, G., Ferres-Marco, D., and Goodman, C.S. (1993). Mutations affecting growth cone guidance in Drosophila: genes necessary for guidance toward or away from the midline. Neuron 10, 409-426.
Skehel PA, Armitage BA, Bartsch D, Hu Y, Kaang BK, Siegelbaum SA, Kandel ER, Martin KC (1995) Proteins functioning in synaptic transmission at the sensory to motor synapse of Aplysia. Neuropharmacology 34, 1379-85.
Skehel, P.A., Fabian-Fine, R., and Kandel, E.R. (2000). Mouse VAP33 is associated with the endoplasmic reticulum and microtubules. Proceedings of the National Academy of Sciences of the United States of America 97, 1101-1106.
Soussan, L., Burakov, D., Daniels, M.P., Toister-Achituv, M., Porat, A., Yarden, Y., and Elazar, Z. (1999). ERG30, a VAP-33-related protein, functions in protein transport mediated by COPI vesicles. The Journal of cell biology 146, 301-311.
Stanley, H., Botas, J., and Malhotra, V. (1997). The mechanism of Golgi segregation during mitosis is cell typespecific. Proceedings of the National Academy of Sciences of the United States of America 94, 14467-14470.
Stefan, C.J., Manford, A.G., Baird, D., Yamada-Hanff, J., Mao, Y., and Emr, S.D. (2011). Osh proteins regulate phosphoinositide metabolism at ER-plasma membrane contact sites. Cell 744, 389-401.
Suchanek, M., Hynynen, R., Wohlfahrt, G., Lehto, M., Johansson, M., Saarinen, H., Radzikowska, A., Thiele, C, and Olkkonen, V.M. (2007). The mammalian oxysterol-binding protein-related proteins (ORPs) bind 25- hydroxycholesterol in an evolutionarily conserved pocket. The Biochemical journal 405, 473-480.
Swanton, C, Marani, M., Pardo, O., Warne, P.H., Kelly, G., Sahai, E., Elustondo, F., Chang, J., Temple, J., Ahmed, A. A., et al. (2007). Regulators of mitotic arrest and ceramide metabolism are determinants of sensitivity to paclitaxel and other chemo therapeutic drugs.
Cancer cell 11, 498-512.
40530616.1 Tanouye, M.A., and Wyman, R.J. (1980). Motor outputs of giant nerve fiber in Drosophila. Journal of neurophysiology 44, 405-421.
Teuling E, Ahmed S, Haasdijk E, Demmers J, Steinmetz MO, Akhmanova A, Jaarsma D, Hoogenraad CC (2007) Motor neuron disease-associated mutant vesicle-associated membrane protein-associated protein (VAP) B recruits wild-type VAPs into endoplasmic reticulum-derived tubular aggregates. J Neurosci 27, 9801-15.
Tobisawa S, Hozumi Y, et al. (2003) Mutant SOD1 linked to familial amyotrophic lateral sclerosis, but not wild- type SOD1, induces ER stress in COS7 cells and transgenic mice. Biochem Biophys Res Commun 303, 496-503.
Tsuda H, Han SM, et al. (2008) The amyotrophic lateral sclerosis 8 protein VAPB is cleaved, secreted, and acts as a ligand for Eph receptors. Cell 133, 963-77.
Van Hoecke A, Schoonaert L, et al. (2012) EPHA4 is a disease modifier of amyotrophic lateral sclerosis in animal models and in humans. Nat Med 18, 1418-22.
Wu CH, Fallini C, et al. (2012) Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis. Nature 488, 499-503.
Wyles JP, McMaster CR, Ridgway ND (2002) Vesicle-associated membrane protein- associated protein-A (VAP-A) interacts with the oxysterol-binding protein to modify export from the endoplasmic reticulum. J Biol Chem 277, 29908-18.
Yang Z, Huh SU, Drennan JM, Kathuria H, Martinez JS, Tsuda H, Hall MC, Clemens JC (2012) Drosophila Vap-33 is required for axonal localization of Dscam isoforms. J Neurosci 32, 17241-50.
Zhou J, Yi J, Fu R, Liu E, Siddique T, Rios E, Deng HX (2010) Hyperactive intracellular calcium signaling associated with localized mitochondrial defects in skeletal muscle of an animal model of amyotrophic lateral sclerosis. J Biol Chem 285, 705-12.
40530616.1

Claims

CLAIMS What is claimed is:
1. A method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of a Major Sperm Protein (MSP) composition or functionally active fragment or derivative thereof to the individual.
2. The method of claim 1, wherein the neurological disorder is
amyotrophic lateral sclerosis (ALS).
3. The method of claim 2, wherein the ALS is familial or sporadic.
4. The method of claim 1, wherein the individual has at least one symptom of ALS.
5. The method of claim 1, wherein the individual is suspected of
having ALS.
6. The method of claim 1, further comprising the step of diagnosing ALS in the individual.
7. The method of claim 1, wherein the MSP composition is provided to the individual multiple times.
8. The method of claim 1, wherein MSP composition is administered to the individual intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intrapleurally, intranasally, intravitreally, intramuscularly, intraperitoneally, subcutaneously, subconjunctivally, intravesicularly, mucosally, intrapericardially, orally, by inhalation,
40530616.1 by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, or via a lavage.
9. The method of claim 1, wherein the composition is provided to the individual in proteinaceous form.
10. The method of claim 1, wherein the composition is provided to the individual in nucleic acid form.
11. The method of claim 10, wherein the nucleic acid is an expression vector that encodes MSP.
12. The method of claim 11, wherein the expression vector is a viral vector.
13. The method of claim 12, wherein the viral vector is an adeno- associated viral vector, an adenoviral vector, a retroviral vector, or a lentiviral vector.
14. The method of claim 1, wherein the individual is provided an
additional therapy for the neurological disorder or therapy for at least one symptom thereof.
15. The method of claim 14, wherein the additional therapy is riluzole.
16. The method of claim 1, wherein the neurological disorder is ALS.
17. A pharmaceutical composition comprising a MSP composition or functionally active fragment or derivative thereof.
18. The composition of claim 17, wherein the MSP composition is in proteinaceous form.
19. The composition of claim 17, wherein the MSP composition is in nucleic acid form.
40530616.1
20. The composition of claim 19, wherein the nucleic acid is an expression vector.
21. The composition of claim 20, wherein the expression vector is a viral vector.
22. The composition of claim 21, wherein the viral vector is an adeno- associated viral vector, an adenoviral vector, a retroviral vector, or a lentiviral vector.
23. The composition of claim 17, wherein the composition comprises one or more modifications.
24. The composition of claim 23, wherein the one or more
modifications extend the half life of the composition.
25. The composition of claim 23, wherein the MSP composition
comprises one or more polyethylene glycol groups.
26. The composition of claim 23, wherein the MSP composition
comprises one or more immunoglobulins.
27. The composition of claim 23, wherein the MSP composition
comprises at least one D amino acid.
28. The composition of claim 23, wherein the MSP composition
comprises a label, tag, or both.
29. The composition of claim 23, wherein the MSP composition is fused in-frame with another polypeptide.
30. The composition of claim 17, wherein the functionally active fragment or derivative thereof is at least 80%, 85%, 90%, 95%, 97%, or 99% identical to SEQ ID NO:2 or SEQ ID NO:26.
40530616.1
31. The composition of claim 17, wherein the functionally active fragment or derivative thereof comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid alterations compared to SEQ ID NO:2 or SEQ ID NO:26.
32. The composition of claim 31, wherein the alteration comprises an amino acid substitution, deletion, addition, or inversion.
33. The composition of claim 17, wherein the functionally active
fragment or derivative thereof is no more than 120, 115, 110, 105, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40 35, 30, or 25 amino acids in length.
34. The composition of claim 17, wherein the functionally active
fragment or derivative thereof comprises at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, or 120 contiguous amino acids of SEQ ID NO:2 or SEQ ID NO:26.
35. The composition of claim 17, wherein the functionally active
fragment or derivative thereof comprises a N-terminal truncation.
36. The composition of claim 35, wherein the N-terminal truncation comprises removal of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
37. The composition of claim 17, wherein the functionally active
fragment or derivative thereof comprises a C-terminal truncation.
38. The composition of claim 37, wherein the C-terminal truncation comprises removal of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33,
40530616.1 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
39. The composition of claim 17, wherein the functionally active
fragment or derivative thereof comprises a N-terminal truncation and a C-terminal truncation.
40. The composition of claim 39, wherein the N-terminal truncation comprises removal of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26 and wherein the C-terminal truncation comprises removal of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acids compared to SEQ ID NO:2 or SEQ ID NO:26.
41. A method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of FOXOl in the individual.
42. The method of claim 41, wherein the agent is a small molecule, nucleic acid, peptide, or protein.
43. The method of claim 42, wherein the nucleic acid is a
polynucleotide that encodes at least part of FOXOl.
44. The method of claim 41, further comprising the step of providing a therapeutically effective amount of a MSP composition or functionally active fragment or derivative thereof to the individual.
40530616.1
45. The method of claim 41, wherein the neurological disorder is ALS.
46. A method of treating, preventing, or reducing the risk of having a neurological disorder in an individual, comprising the step of providing a therapeutically effective amount of one or more agents that increases the expression level and/or activity of Osbp in the individual.
47. The method of claim 46, wherein the agent is a small molecule, nucleic acid, peptide, or protein.
48. The method of claim 47, wherein the nucleic acid is a
polynucleotide that encodes at least part of Osbp.
49. The method of claim 46, further comprising the step of providing a therapeutically effective amount of a MSP composition or functionally active fragment or derivative thereof to the individual.
50. The method of claim 46, wherein the neurological disorder is ALS.
51. The composition of claim 17, wherein the MSP composition
comprises an antibody fragment.
52. The composition of claim 17, wherein the MSP composition
comprises Fc.
53. The composition of claim 17, wherein the MSP composition
comprises collagen.
54. The composition of claim 17, wherein the MSP composition
comprises albumin.
55. The composition of claim 17, wherein the MSP composition is further defined as a MSP multimer.
40530616.1
56. The composition of claim 17, wherein the MSP composition is further defined as a MSP dimer.
57. The composition of claim 17, wherein the MSP composition is further defined as a MSP oligomer.
58. The composition of claim 17, wherein the MSP composition is further defined as a peptide shuffled MSP molecule.
59. A kit comprising the compositon of any one of claims 17-40 and 51-58.
60. The kit of claim 59, further comprising an additional therapy for a neurological disorder or therapy for at least one symptom thereof.
40530616.1
PCT/US2014/030598 2013-03-15 2014-03-17 Msp and its receptors in the therapy of amyotrophic lateral sclerosis WO2014145776A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361801218P 2013-03-15 2013-03-15
US61/801,218 2013-03-15
US201361847889P 2013-07-18 2013-07-18
US61/847,889 2013-07-18

Publications (2)

Publication Number Publication Date
WO2014145776A2 true WO2014145776A2 (en) 2014-09-18
WO2014145776A3 WO2014145776A3 (en) 2015-01-15

Family

ID=51538539

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/030598 WO2014145776A2 (en) 2013-03-15 2014-03-17 Msp and its receptors in the therapy of amyotrophic lateral sclerosis

Country Status (1)

Country Link
WO (1) WO2014145776A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030118556A1 (en) * 2001-12-21 2003-06-26 Kaspar Brian K. Targeted retrograde gene delivery to motor neurons
US6642262B2 (en) * 1998-11-13 2003-11-04 Aventia Pharma S.A. Riluzole and alpha-tocopherol combination
WO2012156351A1 (en) * 2011-05-13 2012-11-22 Vib Vzw Epha4 is a disease modifier in motor neuron disease

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6642262B2 (en) * 1998-11-13 2003-11-04 Aventia Pharma S.A. Riluzole and alpha-tocopherol combination
US20030118556A1 (en) * 2001-12-21 2003-06-26 Kaspar Brian K. Targeted retrograde gene delivery to motor neurons
WO2012156351A1 (en) * 2011-05-13 2012-11-22 Vib Vzw Epha4 is a disease modifier in motor neuron disease

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TSUDA ET AL. THE AMYOTROPHIC LATERAL SCLEROSIS 8 PROTEIN VAPB IS CLEAVED, SECRETED, AND ACTS AS A LIGAND FOR EPH RECEPTORS. vol. 133, no. 6, 13 June 2008, pages 963 - 977 *

Also Published As

Publication number Publication date
WO2014145776A3 (en) 2015-01-15

Similar Documents

Publication Publication Date Title
Buraei et al. The β subunit of voltage-gated Ca2+ channels
Hibino et al. RIM binding proteins (RBPs) couple Rab3-interacting molecules (RIMs) to voltage-gated Ca2+ channels
Laprise et al. The FERM protein Yurt is a negative regulatory component of the Crumbs complex that controls epithelial polarity and apical membrane size
Hollingsworth et al. Defective trafficking of rhodopsin and its role in retinal degenerations
Moustaqim-Barrette et al. The amyotrophic lateral sclerosis 8 protein, VAP, is required for ER protein quality control
CA2917389C (en) Acetylcholinesterase-derived cyclic peptide for treating neurodegenerative disorders
Dason et al. Multiple roles for frequenin/NCS-1 in synaptic function and development
JP2000506392A (en) A novel human metabotropic glutamate receptor
Hsu et al. Multiple domains in the Crumbs Homolog 2a (Crb2a) protein are required for regulating rod photoreceptor size
Hoehne et al. Loss-and gain-of-function analysis of the lipid raft proteins Reggie/Flotillin in Drosophila: they are posttranslationally regulated, and misexpression interferes with wing and eye development
Beck et al. Regulation of Fasciclin II and synaptic terminal development by the splicing factor beag
Ashlin et al. Pitpnc1a regulates zebrafish sleep and wake behavior through modulation of insulin-like growth factor signaling
WO2014145776A2 (en) Msp and its receptors in the therapy of amyotrophic lateral sclerosis
JP2019508018A (en) SorCS peptides and their uses
US8193315B2 (en) Excitatory glycine receptors and methods
Chen et al. Drosophila homolog of the human carpenter syndrome linked gene, MEGF8, is required for synapse development and function
US9253964B2 (en) Mutant alpha-synuclein, and methods using same
Pawlikowski et al. Formation of complex AChR aggregates in vitro requires α‐dystrobrevin
Brija Alternative splicing and RNA editing of the Complexin C-terminus regulates neurotransmitter release in Drosophila
Kiss Genotype-phenotype relationship in Drosophila type IV collagen mutants
Küffer The Role of Axonal Prion Protein in Myelin Maintenance
Beeler-Muscat Characterization of Optimus Prime, A Novel Regulator Of Synaptic Transmission In Drosophila
Royal Combinatorial diversity of two-pore-domain k+ channels and its involvement in migraine
US20030092004A1 (en) Excitatory glycine receptors and methods
Wang The Role of Secretory Pathway in Neuronal Pruning in Drosophila

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14763332

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 14763332

Country of ref document: EP

Kind code of ref document: A2