Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Mol Genet ; 24(3): 773-86, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274782

ABSTRACT

Mutations in the RNA-binding protein FUS/TLS (FUS) have been linked to the neurodegenerative disease amyotrophic lateral sclerosis (ALS). Although predominantly nuclear, this heterogenous nuclear ribonuclear protein (hnRNP) has multiple functions in RNA processing including intracellular trafficking. In ALS, mutant or wild-type (WT) FUS can form neuronal cytoplasmic inclusions. Asymmetric arginine methylation of FUS by the class 1 arginine methyltransferase, protein arginine methyltransferase 1 (PRMT1), regulates nucleocytoplasmic shuttling of FUS. In motor neurons of primary spinal cord cultures, redistribution of endogenous mouse and that of ectopically expressed WT or mutant human FUS to the cytoplasm led to nuclear depletion of PRMT1, abrogating methylation of its nuclear substrates. Specifically, hypomethylation of arginine 3 of histone 4 resulted in decreased acetylation of lysine 9/14 of histone 3 and transcriptional repression. Distribution of neuronal PRMT1 coincident with FUS also was detected in vivo in the spinal cord of FUS(R495X) transgenic mice. However, nuclear PRMT1 was not stable postmortem obviating meaningful evaluation of ALS autopsy cases. This study provides evidence for loss of PRMT1 function as a consequence of cytoplasmic accumulation of FUS in the pathogenesis of ALS, including changes in the histone code regulating gene transcription.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cytoplasm/metabolism , Protein-Arginine N-Methyltransferases/metabolism , RNA-Binding Protein FUS/metabolism , Repressor Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Nucleus/metabolism , Cells, Cultured , DNA Methylation , Disease Models, Animal , Histones/metabolism , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Spinal Cord/metabolism
2.
Cell Stress Chaperones ; 19(3): 421-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24092395

ABSTRACT

Heat shock proteins (HSPs) are attractive therapeutic targets for neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), characterized by aberrant formation of protein aggregates. Although motor neurons have a high threshold for activation of HSP genes, HSP90 inhibitors are effective inducers. This study evaluated NXD30001, a novel, small molecule HSP90 inhibitor based on the radicicol backbone, for its ability to induce neuronal HSPs and for efficacy in an experimental model of ALS based on mutations in superoxide-dismutase 1 (SOD1). In motor neurons of dissociated murine spinal cord cultures, NXD30001-induced expression of HSP70/HSPA1 (iHSP70) and its co-chaperone HSP40/DNAJ through activation of HSF1 and exhibited a protective profile against SOD1(G93A) similar to geldanamycin, but with less toxicity. Treatment prevented protein aggregation, mitochondrial fragmentation, and motor neuron death, important features of mutant SOD1 toxicity, but did not effectively prevent aberrant intracellular Ca(2+) accumulation. NXD30001 distributed to brain and spinal cord of wild-type and SOD1(G93A) transgenic mice following intraperitoneal injection; however, unlike in culture, in vivo levels of SOD1 were not reduced. NXD30001-induced expression of iHSP70 in skeletal and cardiac muscle and, to a lesser extent, in kidney, but not in liver, spinal cord, or brain, with either single or repeated administration. NXD30001 is a very useful experimental tool in culture, but these data point to the complex nature of HSP gene regulation in vivo and the necessity for early evaluation of the efficacy of novel HSP inducers in target tissues in vivo.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Lactones/pharmacology , Nerve Tissue/metabolism , Oximes/pharmacology , Small Molecule Libraries/pharmacology , Animals , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Green Fluorescent Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Homeostasis/drug effects , Inclusion Bodies/metabolism , Lactones/administration & dosage , Lactones/pharmacokinetics , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Dynamics/drug effects , Motor Neurons/drug effects , Motor Neurons/metabolism , Nerve Tissue/drug effects , Oximes/administration & dosage , Oximes/pharmacokinetics , Phosphorylation/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacokinetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tissue Culture Techniques
3.
Hum Mol Genet ; 21(1): 136-49, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21965298

ABSTRACT

Mutations in FUS/TLS (fused in sarcoma/translated in liposarcoma) cause an inheritable form of amyotrophic lateral sclerosis (ALS6). In contrast to FUS(WT), which is concentrated in the nucleus, these mutants are abnormally distributed in the cytoplasm where they form inclusions and associate with stress granules. The data reported herein demonstrate the importance of protein arginine methylation in nuclear-cytoplasmic shuttling of FUS and abnormalities of ALS-causing mutants. Depletion of protein arginine methyltransferase 1 (PRMT1; the enzyme that methylates FUS) in mouse embryonic fibroblasts by gene knockout, or in human HEK293 cells by siRNA knockdown, diminished the ability of ALS-linked FUS mutants to localize to the cytoplasm and form inclusions. To examine properties of FUS mutants in the context of neurons vulnerable to the disease, FUS(WT) and ALS-linked FUS mutants were expressed in motor neurons of dissociated murine spinal cord cultures. In motor neurons, shRNA-mediated PRMT1 knockdown concomitant with the expression of FUS actually accentuated the shift in distribution of ALS-linked FUS mutants from the nucleus to the cytoplasm. However, when PRMT1 was inhibited prior to expression of ALS-linked FUS mutants, by pretreatment with a global methyltransferase inhibitor, ALS-linked FUS mutants were sequestered in the nucleus and cytoplasmic inclusions were reduced, as in the cell lines. Mitochondria were significantly shorter in neurons with cytoplasmic ALS-linked FUS mutants, a factor that could contribute to toxicity. We propose that arginine methylation by PRMT1 participates in the nuclear-cytoplasmic shuttling of FUS, particularly of ALS6-associated mutants, and thus contributes to the toxic gain of function conferred by these disease-causing mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mutation , Protein-Arginine N-Methyltransferases/metabolism , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/metabolism , Repressor Proteins/metabolism , Amino Acid Motifs , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Arginine/metabolism , Cell Line , Cell Nucleus/genetics , Cells, Cultured , Cytoplasm/genetics , Humans , Methylation , Mice , Motor Neurons/metabolism , Protein Transport , Protein-Arginine N-Methyltransferases/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/toxicity , Repressor Proteins/genetics
4.
Neurobiol Dis ; 42(3): 265-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21296666

ABSTRACT

The combination of Ca(2+) influx during neurotransmission and low cytosolic Ca(2+) buffering contributes to the preferential vulnerability of motor neurons in amyotrophic lateral sclerosis (ALS). This study investigated the relationship among Ca(2+) accumulation in intracellular compartments, mitochondrial abnormalities, and protein aggregation in a model of familial ALS (fALS1). Human SOD1, wild type (SOD1(WT)) or with the ALS-causing mutation G93A (SOD1(G93A)), was expressed in motor neurons of dissociated murine spinal cord-dorsal root ganglia (DRG) cultures. Elevation of mitochondrial Ca(2+) ([Ca(2+)](m)), decreased mitochondrial membrane potential (Δψ) and rounding of mitochondria occurred early, followed by increased endoplasmic reticular Ca(2+) ([Ca(2+)](ER)), elevated cytosolic Ca(2+) ([Ca(2+)](c)), and subsequent appearance of SOD1(G93A) inclusions (a consequence of protein aggregation). [Ca(2+)](c) was elevated to a greater extent in neurons with inclusions than in those with diffusely distributed SOD1(G93A) and promoted aggregation of mutant protein, not vice versa: both [Ca(2+)](c) and the percentage of neurons with SOD1(G93A) inclusions were reduced by co-expressing the cytosolic Ca(2+)-buffering protein, calbindin D-28K; treatment with the heat shock protein inducer, geldanamycin, prevented inclusions but not the increase in [Ca(2+)](c), [Ca(2+)](m) or loss of Δψ, and inhibiting proteasome activity with epoxomicin, known to promote aggregation of disease-causing mutant proteins including SOD1(G93A), had no effect on Ca(2+) levels. Both expression of SOD1(G93A) and epoxomicin-induced inhibition of proteasome activity caused mitochondrial rounding, independent of Ca(2+) dysregulation and reduced Δψ. That geldanamycin prevented inclusions and mitochondrial rounding, but not Ca(2+) dysregulation or loss of Δψ indicates that chaperone-based therapies to prevent protein aggregation may require co-therapy to address these other underlying mechanisms of toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Calcium/metabolism , Mitochondria/metabolism , Motor Neurons/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cells, Cultured , Disease Models, Animal , Membrane Potential, Mitochondrial/physiology , Mice , Microscopy, Confocal , Mitochondria/pathology , Motor Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
5.
Neuroreport ; 21(15): 976-9, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20736867

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset, rapidly progressing, fatal disease occurring in both familial and sporadic forms. Mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1) cause ALS through a gain of toxic function. Calpain activity is increased in mutant SOD1 (SOD1(G93A)) transgenic mice and in models of ischemia because of increased cytosolic calcium, which has been documented in motor neurons in rodent models of familial ALS and in sporadic ALS patients. We report that inhibition of calpain activity using calpastatin prevented the toxicity of SOD1(G93A) in motor neurons of dissociated spinal cord cultures, prolonging viability of and reducing the proportion containing SOD1(G93A) inclusions. The data support the central role of calcium dysregulation in ALS and identify a potential therapeutic pathway.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Calcium-Binding Proteins/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Motor Neurons/drug effects , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Calpain/antagonists & inhibitors , Cell Aggregation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Transfer Techniques , Mice , Motor Neurons/pathology
6.
Hum Mol Genet ; 19(4): 671-83, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19959528

ABSTRACT

TDP-43 has been found in inclusion bodies of multiple neurological disorders, including amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson's disease and Alzheimer's disease. Mutations in the TDP-43 encoding gene, TARDBP, have been subsequently reported in sporadic and familial ALS patients. In order to investigate the pathogenic nature of these mutants, the effects of three consistently reported TARDBP mutations (A315T, G348C and A382T) were tested in cell lines, primary cultured motor neurons and living zebrafish embryos. Each of the three mutants and wild-type (WT) human TDP-43 localized to nuclei when expressed in COS1 and Neuro2A cells by transient transfection. However, when expressed in motor neurons from dissociated spinal cord cultures these mutant TARDBP alleles, but less so for WT TARDBP, were neurotoxic, concomitant with perinuclear localization and aggregation of TDP-43. Finally, overexpression of mutant, but less so of WT, human TARDBP caused a motor phenotype in zebrafish (Danio rerio) embryos consisting of shorter motor neuronal axons, premature and excessive branching as well as swimming deficits. Interestingly, knock-down of zebrafisfh tardbp led to a similar phenotype, which was rescued by co-expressing WT but not mutant human TARDBP. Together these approaches showed that TARDBP mutations cause motor neuron defects and toxicity, suggesting that both a toxic gain of function as well as a novel loss of function may be involved in the molecular mechanism by which mutant TDP-43 contributes to disease pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Motor Activity , Mutation , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Genetically Modified , Cell Line , Cells, Cultured , Humans , Mice , Motor Neurons/metabolism , Zebrafish/genetics , Zebrafish/physiology
7.
J Neuropathol Exp Neurol ; 68(6): 642-52, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19458545

ABSTRACT

Mutations in NEFL encoding the light neurofilament subunit (NFL) cause Charcot-Marie-Tooth disease type 2E (CMT2E), which affects both motor and sensory neurons. We expressed the disease-causing mutants NFL and NFL in motor neurons of dissociated spinal cord-dorsal root ganglia and demonstrated that they are incorporated into the preexisting neurofilament network but eventually disrupt neurofilaments without causing significant motor neuron death. Importantly, rounding of mitochondria and reduction in axonal diameter occurred before disruption of the neurofilament network, indicating that mitochondrial dysfunction contributes to the pathogenesis of CMT2E, as well as to CMT caused by mitofusin mutations. Heat shock proteins (HSPs) are involved in the formation of the neurofilament network and in protecting cells from misfolded mutant proteins. Cotransfection of HSPB1 with mutated NEFL maintained the neurofilament network, axonal diameter, and mitochondrial length in motor neurons expressing NFL, but not NFL. Conversely, HSPA1 cotransfection was effective in motor neurons expressing NFL, but not NFL. Thus, there are NFL mutant-specific differences in the ability of individual HSPs to prevent neurofilament abnormalities, reduction in axonal caliber, and disruption of mitochondrial morphology in motor neurons. These results suggest that HSP inducers have therapeutic potential for CMT2E but that their efficacy would depend on the profile of HSPs induced and the type of NEFL mutation.


Subject(s)
Axons/pathology , Heat-Shock Proteins/metabolism , Mitochondria/pathology , Motor Neurons/ultrastructure , Mutation , Neoplasm Proteins/metabolism , Neurofilament Proteins/metabolism , Amino Acids/genetics , Analysis of Variance , Animals , Arginine/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Ganglia, Spinal/cytology , Glutamine/genetics , Green Fluorescent Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Mice , Microinjections/methods , Molecular Chaperones , Motor Neurons/pathology , Neoplasm Proteins/genetics , Neurofilament Proteins/genetics , Proline/genetics , Spinal Cord/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods
8.
Cell Stress Chaperones ; 12(2): 151-62, 2007.
Article in English | MEDLINE | ID: mdl-17688194

ABSTRACT

Induction of heat shock proteins (Hsps) is under investigation as treatment for neurodegenerative disorders, yet many types of neurons, including motor neurons that degenerate in amyotrophic lateral sclerosis (ALS), have a high threshold for activation of the major transcription factor mediating stress-induced Hsp upregulation, heat shock transcription factor 1 (Hsf1). Hsf1 is tightly regulated by a series of inhibitory checkpoints that include sequestration in multichaperone complexes governed by Hsp90. This study examined the role of multichaperone complexes in governing the heat shock response in motor neurons. Hsp90 inhibitors induced expression of Hsp70 and Hsp40 and transactivation of a human inducible hsp70 promoter-green fluorescent protein (GFP) reporter construct in motor neurons of dissociated spinal cord-dorsal root ganglion (DRG) cultures. On the other hand, overexpression of activator of Hsp90 adenosine triphosphatase ([ATPase 1], Aha1), which should mobilize Hsf1 by accelerating turnover of mature, adenosine triphosphate-(ATP) bound Hsp90 complexes, and death domain-associated protein (Daxx), which in cell lines has been shown to promote transcription of heat shock genes by relieving inhibition exerted by interactions between nuclear Hsp90/multichaperone complexes and trimeric Hsf1, failed to induce Hsps in the absence or presence of heat shock. These results indicate that disruption of multichaperone complexes alone is not sufficient to activate the neuronal heat shock response. Furthermore, in motor neurons, induction of Hsp70 by Hsp90-inhibiting drugs was prevented by overexpression of wild-type Hsfl, contrary to what would be expected for a classical Hsf1-mediated pathway. These results point to additional differences in regulation of hsp genes in neuronal and nonneuronal cells.


Subject(s)
DNA-Binding Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Motor Neurons/metabolism , Transcription Factors/metabolism , Animals , Carrier Proteins/metabolism , Co-Repressor Proteins , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heat Shock Transcription Factors , Humans , Hyperthermia, Induced , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Chaperones , Motor Neurons/cytology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...