Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38895337

ABSTRACT

RNA binding proteins have emerged as central players in the mechanisms of many neurodegenerative diseases. In particular, a proteinopathy of fu sed in s arcoma (FUS) is present in some instances of familial Amyotrophic lateral sclerosis (ALS) and about 10% of sporadic FTLD. Here we establish that focal injection of sonicated human FUS fibrils into brains of mice in which ALS-linked mutant or wild-type human FUS replaces endogenous mouse FUS is sufficient to induce focal cytoplasmic mislocalization and aggregation of mutant and wild-type FUS which with time spreads to distal regions of the brain. Human FUS fibril-induced FUS aggregation in the mouse brain of humanized FUS mice is accelerated by an ALS-causing FUS mutant relative to wild-type human FUS. Injection of sonicated human FUS fibrils does not induce FUS aggregation and subsequent spreading after injection into naïve mouse brains containing only mouse FUS, indicating a species barrier to human FUS aggregation and its prion-like spread. Fibril-induced human FUS aggregates recapitulate pathological features of FTLD including increased detergent insolubility of FUS and TAF15 and amyloid-like, cytoplasmic deposits of FUS that accumulate ubiquitin and p62, but not TDP-43. Finally, injection of sonicated FUS fibrils is shown to exacerbate age-dependent cognitive and behavioral deficits from mutant human FUS expression. Thus, focal seeded aggregation of FUS and further propagation through prion-like spread elicits FUS-proteinopathy and FTLD-like disease progression.

2.
Mol Neurodegener ; 19(1): 46, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862967

ABSTRACT

RNA binding proteins have emerged as central players in the mechanisms of many neurodegenerative diseases. In particular, a proteinopathy of fused in sarcoma (FUS) is present in some instances of familial Amyotrophic lateral sclerosis (ALS) and about 10% of sporadic Frontotemporal lobar degeneration (FTLD). Here we establish that focal injection of sonicated human FUS fibrils into brains of mice in which ALS-linked mutant or wild-type human FUS replaces endogenous mouse FUS is sufficient to induce focal cytoplasmic mislocalization and aggregation of mutant and wild-type FUS which with time spreads to distal regions of the brain. Human FUS fibril-induced FUS aggregation in the mouse brain of humanized FUS mice is accelerated by an ALS-causing FUS mutant relative to wild-type human FUS. Injection of sonicated human FUS fibrils does not induce FUS aggregation and subsequent spreading after injection into naïve mouse brains containing only mouse FUS, indicating a species barrier to human FUS aggregation and its prion-like spread. Fibril-induced human FUS aggregates recapitulate pathological features of FTLD including increased detergent insolubility of FUS and TAF15 and amyloid-like, cytoplasmic deposits of FUS that accumulate ubiquitin and p62, but not TDP-43. Finally, injection of sonicated FUS fibrils is shown to exacerbate age-dependent cognitive and behavioral deficits from mutant human FUS expression. Thus, focal seeded aggregation of FUS and further propagation through prion-like spread elicits FUS-proteinopathy and FTLD-like disease progression.


Subject(s)
Disease Progression , Frontotemporal Dementia , Mice, Transgenic , RNA-Binding Protein FUS , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/genetics , Protein Aggregation, Pathological/metabolism , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics
3.
Nat Neurosci ; 27(1): 34-47, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37996528

ABSTRACT

The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , Axons/physiology , Denervation , DNA-Binding Proteins/genetics , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Motor Neurons/metabolism , Stathmin/genetics , Stathmin/metabolism
4.
Proc Natl Acad Sci U S A ; 119(49): e2123487119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36454749

ABSTRACT

Hexanucleotide G4C2 repeat expansions in the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Dipeptide repeat proteins (DPRs) generated by translation of repeat-containing RNAs show toxic effects in vivo as well as in vitro and are key targets for therapeutic intervention. We generated human antibodies that bind DPRs with high affinity and specificity. Anti-GA antibodies engaged extra- and intra-cellular poly-GA and reduced aggregate formation in a poly-GA overexpressing human cell line. However, antibody treatment in human neuronal cultures synthesizing exogenous poly-GA resulted in the formation of large extracellular immune complexes and did not affect accumulation of intracellular poly-GA aggregates. Treatment with antibodies was also shown to directly alter the morphological and biochemical properties of poly-GA and to shift poly-GA/antibody complexes to more rapidly sedimenting ones. These alterations were not observed with poly-GP and have important implications for accurate measurement of poly-GA levels including the need to evaluate all centrifugation fractions and disrupt the interaction between treatment antibodies and poly-GA by denaturation. Targeting poly-GA and poly-GP in two mouse models expressing G4C2 repeats by systemic antibody delivery for up to 16 mo was well-tolerated and led to measurable brain penetration of antibodies. Long-term treatment with anti-GA antibodies produced improvement in an open-field movement test in aged C9orf72450 mice. However, chronic administration of anti-GA antibodies in AAV-(G4C2)149 mice was associated with increased levels of poly-GA detected by immunoassay and did not significantly reduce poly-GA aggregates or alleviate disease progression in this model.


Subject(s)
Genes, Regulator , Poly A , Animals , Humans , Mice , Antigen-Antibody Complex , C9orf72 Protein/genetics , Dipeptides , Disease Models, Animal
5.
Mol Neurodegener ; 16(1): 61, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488813

ABSTRACT

Mutations in FUS, an RNA-binding protein involved in multiple steps of RNA metabolism, are associated with the most severe forms of amyotrophic lateral sclerosis (ALS). Accumulation of cytoplasmic FUS is likely to be a major culprit in the toxicity of FUS mutations. Thus, preventing cytoplasmic mislocalization of the FUS protein may represent a valuable therapeutic strategy. FUS binds to its own pre-mRNA creating an autoregulatory loop efficiently buffering FUS excess through multiple proposed mechanisms including retention of introns 6 and/or 7. Here, we introduced a wild-type FUS gene allele, retaining all intronic sequences, in mice whose heterozygous or homozygous expression of a cytoplasmically retained FUS protein (Fus∆NLS) was previously shown to provoke ALS-like disease or postnatal lethality, respectively. Wild-type FUS completely rescued the early lethality caused by the two Fus∆NLS alleles, and improved the age-dependent motor deficits and reduced lifespan caused by heterozygous expression of mutant FUS∆NLS. Mechanistically, wild-type FUS decreased the load of cytoplasmic FUS, increased retention of introns 6 and 7 in the endogenous mouse Fus mRNA, and decreased expression of the mutant mRNA. Thus, the wild-type FUS allele activates the homeostatic autoregulatory loop, maintaining constant FUS levels and decreasing the mutant protein in the cytoplasm. These results provide proof of concept that an autoregulatory competent wild-type FUS expression could protect against this devastating, currently intractable, neurodegenerative disease.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Disease Models, Animal , Gene Expression Regulation/genetics , RNA-Binding Protein FUS/physiology , Alleles , Amyotrophic Lateral Sclerosis/genetics , Animals , Cytoplasm/metabolism , Frontotemporal Dementia/genetics , Genes, Lethal , Genetic Complementation Test , Humans , Introns/genetics , Mice , Mice, Transgenic , Mutation , Protein Binding , RNA Precursors/metabolism , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , Recombinant Proteins/metabolism , Transgenes
6.
Nat Neurosci ; 24(8): 1089-1099, 2021 08.
Article in English | MEDLINE | ID: mdl-34083786

ABSTRACT

Methods to enhance adult neurogenesis by reprogramming glial cells into neurons enable production of new neurons in the adult nervous system. Development of therapeutically viable approaches to induce new neurons is now required to bring this concept to clinical application. Here, we successfully generate new neurons in the cortex and dentate gyrus of the aged adult mouse brain by transiently suppressing polypyrimidine tract binding protein 1 using an antisense oligonucleotide delivered by a single injection into cerebral spinal fluid. Radial glial-like cells and other GFAP-expressing cells convert into new neurons that, over a 2-month period, acquire mature neuronal character in a process mimicking normal neuronal maturation. The new neurons functionally integrate into endogenous circuits and modify mouse behavior. Thus, generation of new neurons in the dentate gyrus of the aging brain can be achieved with a therapeutically feasible approach, thereby opening prospects for production of neurons to replace those lost to neurodegenerative disease.


Subject(s)
Dentate Gyrus , Ependymoglial Cells , Neurogenesis/physiology , Neurons , Polypyrimidine Tract-Binding Protein/antagonists & inhibitors , Animals , Cellular Reprogramming/physiology , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Mice , Neurons/cytology , Neurons/physiology , Oligonucleotides, Antisense
8.
Nat Neurosci ; 23(5): 615-624, 2020 05.
Article in English | MEDLINE | ID: mdl-32284607

ABSTRACT

Hexanucleotide expansions in C9orf72, which encodes a predicted guanine exchange factor, are the most frequent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although repeat expansion has been established to generate toxic products, mRNAs encoding the C9ORF72 protein are also reduced in affected individuals. In this study, we tested how C9ORF72 protein levels affected repeat-mediated toxicity. In somatic transgenic mice expressing 66 GGGGCC repeats, inactivation of one or both endogenous C9orf72 alleles provoked or accelerated, respectively, early death. In mice expressing a C9orf72 transgene with 450 repeats that did not encode the C9ORF72 protein, inactivation of one or both endogenous C9orf72 alleles exacerbated cognitive deficits, hippocampal neuron loss, glial activation and accumulation of dipeptide-repeat proteins from translation of repeat-containing RNAs. Reduced C9ORF72 was shown to suppress repeat-mediated elevation in autophagy. These efforts support a disease mechanism in ALS/FTD resulting from reduced C9ORF72, which can lead to autophagy deficits, synergizing with repeat-dependent gain of toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Animals , DNA Repeat Expansion/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Nat Med ; 26(1): 118-130, 2020 01.
Article in English | MEDLINE | ID: mdl-31873312

ABSTRACT

Gene silencing with virally delivered shRNA represents a promising approach for treatment of inherited neurodegenerative disorders. In the present study we develop a subpial technique, which we show in adult animals successfully delivers adeno-associated virus (AAV) throughout the cervical, thoracic and lumbar spinal cord, as well as brain motor centers. One-time injection at cervical and lumbar levels just before disease onset in mice expressing a familial amyotrophic lateral sclerosis (ALS)-causing mutant SOD1 produces long-term suppression of motoneuron disease, including near-complete preservation of spinal α-motoneurons and muscle innervation. Treatment after disease onset potently blocks progression of disease and further α-motoneuron degeneration. A single subpial AAV9 injection in adult pigs or non-human primates using a newly designed device produces homogeneous delivery throughout the cervical spinal cord white and gray matter and brain motor centers. Thus, spinal subpial delivery in adult animals is highly effective for AAV-mediated gene delivery throughout the spinal cord and supraspinal motor centers.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Dependovirus/metabolism , Gene Silencing , Gene Transfer Techniques , Motor Neurons/pathology , Nerve Degeneration/therapy , Pia Mater/pathology , Spinal Cord/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Atrophy , Disease Progression , Evoked Potentials, Motor , Female , Gene Expression Regulation , Humans , Inflammation/pathology , Interneurons/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle Development , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Pia Mater/physiopathology , Primates , Protein Folding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , Spinal Cord/diagnostic imaging , Spinal Cord/physiopathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Swine
10.
JCI Insight ; 4(10)2019 05 16.
Article in English | MEDLINE | ID: mdl-31092730

ABSTRACT

The discovery of novel biomarkers has emerged as a critical need for therapeutic development in amyotrophic lateral sclerosis (ALS). For some subsets of ALS, such as the genetic superoxide dismutase 1 (SOD1) form, exciting new treatment strategies, such as antisense oligonucleotide-mediated (ASO-mediated) SOD1 silencing, are being tested in clinical trials, so the identification of pharmacodynamic biomarkers for therapeutic monitoring is essential. We identify increased levels of a 7-amino acid endogenous peptide of SOD1 in cerebrospinal fluid (CSF) of human SOD1 mutation carriers but not in other neurological cases or nondiseased controls. Levels of peptide elevation vary based on the specific SOD1 mutation (ranging from 1.1-fold greater than control in D90A to nearly 30-fold greater in V148G) and correlate with previously published measurements of SOD1 stability. Using a mass spectrometry-based method (liquid chromatography-mass spectrometry), we quantified peptides in both extracellular samples (CSF) and intracellular samples (spinal cord from rat) to demonstrate that the peptide distinguishes mutation-specific differences in intracellular SOD1 degradation. Furthermore, 80% and 63% reductions of the peptide were measured in SOD1G93A and SOD1H46R rat CSF samples, respectively, following treatment with ASO, with an improved correlation to mRNA levels in spinal cords compared with the ELISA measuring intact SOD1 protein. These data demonstrate the potential of this peptide as a pharmacodynamic biomarker.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Biomarkers , Peptides/pharmacology , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/therapy , Animals , Biomarkers/cerebrospinal fluid , Disease Models, Animal , Gene Silencing , Humans , Mutation , Peptides/cerebrospinal fluid , Rats , Spinal Cord
11.
Elife ; 82019 02 12.
Article in English | MEDLINE | ID: mdl-30747709

ABSTRACT

Mutations in coding and non-coding regions of FUS cause amyotrophic lateral sclerosis (ALS). The latter mutations may exert toxicity by increasing FUS accumulation. We show here that broad expression within the nervous system of wild-type or either of two ALS-linked mutants of human FUS in mice produces progressive motor phenotypes accompanied by characteristic ALS-like pathology. FUS levels are autoregulated by a mechanism in which human FUS downregulates endogenous FUS at mRNA and protein levels. Increasing wild-type human FUS expression achieved by saturating this autoregulatory mechanism produces a rapidly progressive phenotype and dose-dependent lethality. Transcriptome analysis reveals mis-regulation of genes that are largely not observed upon FUS reduction. Likely mechanisms for FUS neurotoxicity include autophagy inhibition and defective RNA metabolism. Thus, our results reveal that overriding FUS autoregulation will trigger gain-of-function toxicity via altered autophagy-lysosome pathway and RNA metabolism function, highlighting a role for protein and RNA dyshomeostasis in FUS-mediated toxicity.


Subject(s)
Autophagy , Homeostasis , Lysosomes/metabolism , RNA-Binding Protein FUS/biosynthesis , RNA-Binding Protein FUS/toxicity , RNA/metabolism , Animals , Gene Expression Profiling , Humans , Mice, Inbred C57BL , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Mutant Proteins/toxicity , RNA-Binding Protein FUS/genetics
12.
Neuron ; 100(4): 816-830.e7, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30344044

ABSTRACT

Through the generation of humanized FUS mice expressing full-length human FUS, we identify that when expressed at near endogenous murine FUS levels, both wild-type and ALS-causing and frontotemporal dementia (FTD)-causing mutations complement the essential function(s) of murine FUS. Replacement of murine FUS with mutant, but not wild-type, human FUS causes stress-mediated induction of chaperones, decreased expression of ion channels and transporters essential for synaptic function, and reduced synaptic activity without loss of nuclear FUS or its cytoplasmic aggregation. Most strikingly, accumulation of mutant human FUS is shown to activate an integrated stress response and to inhibit local, intra-axonal protein synthesis in hippocampal neurons and sciatic nerves. Collectively, our evidence demonstrates that human ALS/FTD-linked mutations in FUS induce a gain of toxicity that includes stress-mediated suppression in intra-axonal translation, synaptic dysfunction, and progressive age-dependent motor and cognitive disease without cytoplasmic aggregation, altered nuclear localization, or aberrant splicing of FUS-bound pre-mRNAs. VIDEO ABSTRACT.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Axons/physiology , Frontotemporal Dementia/genetics , Loss of Function Mutation/genetics , Protein Biosynthesis/physiology , RNA-Binding Protein FUS/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Cells, Cultured , Female , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , RNA-Binding Protein FUS/biosynthesis
13.
Acta Neuropathol ; 134(1): 97-111, 2017 07.
Article in English | MEDLINE | ID: mdl-28247063

ABSTRACT

A common feature of inherited and sporadic ALS is accumulation of abnormal proteinaceous inclusions in motor neurons and glia. SOD1 is the major protein component accumulating in patients with SOD1 mutations, as well as in mutant SOD1 mouse models. ALS-linked mutations of SOD1 have been shown to increase its propensity to misfold and/or aggregate. Antibodies specific for monomeric or misfolded SOD1 have detected misfolded SOD1 accumulating predominantly in spinal cord motor neurons of ALS patients with SOD1 mutations. We now use seven different conformationally sensitive antibodies to misfolded human SOD1 (including novel high affinity antibodies currently in pre-clinical development) coupled with immunohistochemistry, immunofluorescence and immunoprecipitation to test for the presence of misfolded SOD1 in high quality human autopsy samples. Whereas misfolded SOD1 is readily detectable in samples from patients with SOD1 mutations, it is below detection limits for all of our measures in spinal cord and cortex tissues from patients with sporadic or non-SOD1 inherited ALS. The absence of evidence for accumulated misfolded SOD1 supports a conclusion that SOD1 misfolding is not a primary component of sporadic ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice, Transgenic , Middle Aged , Protein Folding , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Young Adult
14.
Acta Neuropathol ; 133(6): 907-922, 2017 06.
Article in English | MEDLINE | ID: mdl-28357566

ABSTRACT

Mutations in TDP-43 cause amyotrophic lateral sclerosis (ALS), a fatal paralytic disease characterized by degeneration and premature death of motor neurons. The contribution of mutant TDP-43-mediated damage within motor neurons was evaluated using mice expressing a conditional allele of an ALS-causing TDP-43 mutant (Q331K) whose broad expression throughout the central nervous system mimics endogenous TDP-43. TDP-43Q331K mice develop age- and mutant-dependent motor deficits from degeneration and death of motor neurons. Cre-recombinase-mediated excision of the TDP-43Q331K gene from motor neurons is shown to delay onset of motor symptoms and appearance of TDP-43-mediated aberrant nuclear morphology, and abrogate subsequent death of motor neurons. However, reduction of mutant TDP-43 selectively in motor neurons did not prevent age-dependent degeneration of axons and neuromuscular junction loss, nor did it attenuate astrogliosis or microgliosis. Thus, disease mechanism is non-cell autonomous with mutant TDP-43 expressed in motor neurons determining disease onset but progression defined by mutant acting within other cell types.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Motor Neurons/metabolism , Adult , Aged, 80 and over , Aging/metabolism , Aging/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Disease Progression , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Activity/physiology , Motor Neurons/pathology , Mutation , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Young Adult
15.
Neuron ; 90(3): 535-50, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27112497

ABSTRACT

Hexanucleotide expansions in C9ORF72 are the most frequent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Disease mechanisms were evaluated in mice expressing C9ORF72 RNAs with up to 450 GGGGCC repeats or with one or both C9orf72 alleles inactivated. Chronic 50% reduction of C9ORF72 did not provoke disease, while its absence produced splenomegaly, enlarged lymph nodes, and mild social interaction deficits, but not motor dysfunction. Hexanucleotide expansions caused age-, repeat-length-, and expression-level-dependent accumulation of RNA foci and dipeptide-repeat proteins synthesized by AUG-independent translation, accompanied by loss of hippocampal neurons, increased anxiety, and impaired cognitive function. Single-dose injection of antisense oligonucleotides (ASOs) that target repeat-containing RNAs but preserve levels of mRNAs encoding C9ORF72 produced sustained reductions in RNA foci and dipeptide-repeat proteins, and ameliorated behavioral deficits. These efforts identify gain of toxicity as a central disease mechanism caused by repeat-expanded C9ORF72 and establish the feasibility of ASO-mediated therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Frontotemporal Dementia/drug therapy , Guanine Nucleotide Exchange Factors/genetics , Oligonucleotides, Antisense/pharmacology , RNA/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein , DNA Repeat Expansion/genetics , Frontotemporal Dementia/genetics , Mice, Transgenic , Neurons/metabolism , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/genetics
16.
Proc Natl Acad Sci U S A ; 112(50): E6993-7002, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621731

ABSTRACT

Ubiquitous expression of amyotrophic lateral sclerosis (ALS)-causing mutations in superoxide dismutase 1 (SOD1) provokes noncell autonomous paralytic disease. By combining ribosome affinity purification and high-throughput sequencing, a cascade of mutant SOD1-dependent, cell type-specific changes are now identified. Initial mutant-dependent damage is restricted to motor neurons and includes synapse and metabolic abnormalities, endoplasmic reticulum (ER) stress, and selective activation of the PRKR-like ER kinase (PERK) arm of the unfolded protein response. PERK activation correlates with what we identify as a naturally low level of ER chaperones in motor neurons. Early changes in astrocytes occur in genes that are involved in inflammation and metabolism and are targets of the peroxisome proliferator-activated receptor and liver X receptor transcription factors. Dysregulation of myelination and lipid signaling pathways and activation of ETS transcription factors occur in oligodendrocytes only after disease initiation. Thus, pathogenesis involves a temporal cascade of cell type-selective damage initiating in motor neurons, with subsequent damage within glia driving disease propagation.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Expression Profiling , Motor Neurons/metabolism , Mutation , Neuroglia/metabolism , Protein Biosynthesis , Superoxide Dismutase/genetics , Aged , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Humans , Mice , Motor Neurons/pathology , Neuroglia/pathology , Superoxide Dismutase-1
17.
Neuron ; 86(1): 218-32, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25801706

ABSTRACT

Mutations in superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by loss of motor neurons and accompanied by accumulation of misfolded SOD1 onto the cytoplasmic faces of intracellular organelles, including mitochondria and the endoplasmic reticulum (ER). Using inhibition of misfolded SOD1 deposition onto mitochondria as an assay, a chaperone activity abundant in nonneuronal tissues is now purified and identified to be the multifunctional macrophage migration inhibitory factor (MIF), whose activities include an ATP-independent protein folding chaperone. Purified MIF is shown to directly inhibit mutant SOD1 misfolding. Elevating MIF in neuronal cells suppresses accumulation of misfolded SOD1 and its association with mitochondria and the ER and extends survival of mutant SOD1-expressing motor neurons. Accumulated MIF protein is identified to be low in motor neurons, implicating correspondingly low chaperone activity as a component of vulnerability to mutant SOD1 misfolding and supporting therapies to enhance intracellular MIF chaperone activity.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Protein Folding , Superoxide Dismutase/metabolism , Acid Phosphatase/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Endoplasmic Reticulum/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells , Isoenzymes/genetics , Liver/metabolism , Liver/ultrastructure , Mice , Mice, Transgenic , Mitochondria/metabolism , Motor Neurons/physiology , Mutation/genetics , Nerve Tissue Proteins/metabolism , Protein Transport/genetics , Rats , Rats, Transgenic , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tartrate-Resistant Acid Phosphatase
18.
Proc Natl Acad Sci U S A ; 110(46): E4385-92, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24170856

ABSTRACT

Accumulating evidence from mice expressing ALS-causing mutations in superoxide dismutase (SOD1) has implicated pathological immune responses in motor neuron degeneration. This includes microglial activation, lymphocyte infiltration, and the induction of C1q, the initiating component of the classic complement system that is the protein-based arm of the innate immune response, in motor neurons of multiple ALS mouse models expressing dismutase active or inactive SOD1 mutants. Robust induction early in disease course is now identified for multiple complement components (including C1q, C4, and C3) in spinal cords of SOD1 mutant-expressing mice, consistent with initial intraneuronal C1q induction, followed by global activation of the complement pathway. We now test if this activation is a mechanistic contributor to disease. Deletion of the C1q gene in mice expressing an ALS-causing mutant in SOD1 to eliminate C1q induction, and complement cascade activation that follows from it, is demonstrated to produce changes in microglial morphology accompanied by enhanced loss, not retention, of synaptic densities during disease. C1q-dependent synaptic loss is shown to be especially prominent for cholinergic C-bouton nerve terminal input onto motor neurons in affected C1q-deleted SOD1 mutant mice. Nevertheless, overall onset and progression of disease are unaffected in C1q- and C3-deleted ALS mice, thus establishing that C1q induction and classic or alternative complement pathway activation do not contribute significantly to SOD1 mutant-mediated ALS pathogenesis in mice.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Complement C1q/metabolism , Complement Pathway, Classical/immunology , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Animals , Complement C1q/genetics , Gene Deletion , Immunohistochemistry , Mice , Mice, Knockout , Microglia/cytology , Motor Neurons/immunology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Survival Analysis
19.
Proc Natl Acad Sci U S A ; 110(47): E4530-9, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24170860

ABSTRACT

Expanded hexanucleotide repeats in the chromosome 9 open reading frame 72 (C9orf72) gene are the most common genetic cause of ALS and frontotemporal degeneration (FTD). Here, we identify nuclear RNA foci containing the hexanucleotide expansion (GGGGCC) in patient cells, including white blood cells, fibroblasts, glia, and multiple neuronal cell types (spinal motor, cortical, hippocampal, and cerebellar neurons). RNA foci are not present in sporadic ALS, familial ALS/FTD caused by other mutations (SOD1, TDP-43, or tau), Parkinson disease, or nonneurological controls. Antisense oligonucleotides (ASOs) are identified that reduce GGGGCC-containing nuclear foci without altering overall C9orf72 RNA levels. By contrast, siRNAs fail to reduce nuclear RNA foci despite marked reduction in overall C9orf72 RNAs. Sustained ASO-mediated lowering of C9orf72 RNAs throughout the CNS of mice is demonstrated to be well tolerated, producing no behavioral or pathological features characteristic of ALS/FTD and only limited RNA expression alterations. Genome-wide RNA profiling identifies an RNA signature in fibroblasts from patients with C9orf72 expansion. ASOs targeting sense strand repeat-containing RNAs do not correct this signature, a failure that may be explained, at least in part, by discovery of abundant RNA foci with C9orf72 repeats transcribed in the antisense (GGCCCC) direction, which are not affected by sense strand-targeting ASOs. Taken together, these findings support a therapeutic approach by ASO administration to reduce hexanucleotide repeat-containing RNAs and raise the potential importance of targeting expanded RNAs transcribed in both directions.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , DNA Repeat Expansion/genetics , Frontotemporal Lobar Degeneration/drug therapy , Genetic Therapy/methods , Oligonucleotides, Antisense/pharmacology , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Blotting, Southern , C9orf72 Protein , Central Nervous System/cytology , Central Nervous System/metabolism , DNA Primers/genetics , Fibroblasts/metabolism , Frontotemporal Lobar Degeneration/genetics , Genotype , In Situ Hybridization, Fluorescence , Mice , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
20.
J Neurosci ; 33(11): 4657-71, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486940

ABSTRACT

Mitochondria have been proposed as targets for toxicity in amyotrophic lateral sclerosis (ALS), a progressive, fatal adult-onset neurodegenerative disorder characterized by the selective loss of motor neurons. A decrease in the capacity of spinal cord mitochondria to buffer calcium (Ca(2+)) has been observed in mice expressing ALS-linked mutants of SOD1 that develop motor neuron disease with many of the key pathological hallmarks seen in ALS patients. In mice expressing three different ALS-causing SOD1 mutants, we now test the contribution of the loss of mitochondrial Ca(2+)-buffering capacity to disease mechanism(s) by eliminating ubiquitous expression of cyclophilin D, a critical regulator of Ca(2+)-mediated opening of the mitochondrial permeability transition pore that determines mitochondrial Ca(2+) content. A chronic increase in mitochondrial buffering of Ca(2+) in the absence of cyclophilin D was maintained throughout disease course and was associated with improved mitochondrial ATP synthesis, reduced mitochondrial swelling, and retention of normal morphology. This was accompanied by an attenuation of glial activation, reduction in levels of misfolded SOD1 aggregates in the spinal cord, and a significant suppression of motor neuron death throughout disease. Despite this, muscle denervation, motor axon degeneration, and disease progression and survival were unaffected, thereby eliminating mutant SOD1-mediated loss of mitochondrial Ca(2+) buffering capacity, altered mitochondrial morphology, motor neuron death, and misfolded SOD1 aggregates, as primary contributors to disease mechanism for fatal paralysis in these models of familial ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Calcium/metabolism , Mitochondria/metabolism , Motor Neurons/pathology , Superoxide Dismutase/metabolism , Adenosine Triphosphate/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies/pharmacology , Axons/pathology , Calcium-Binding Proteins/metabolism , Cell Death/genetics , Chromatography, Gel , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Hand Strength/physiology , Humans , Immunoprecipitation , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/pathology , Motor Neurons/ultrastructure , Mutation/genetics , Neuromuscular Junction/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/immunology , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL
...