Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
2.
Neurotherapeutics ; 18(2): 1095-1112, 2021 04.
Article in English | MEDLINE | ID: mdl-33786804

ABSTRACT

Vascular dementia is one of the most common forms of dementia in aging population. However, the molecular mechanisms involved in development of disease and the link between the cerebrovascular pathology and the cognitive impairments remain elusive. Currently, one common and/or converging neuropathological pathway leading to dementia is the mislocalization and altered functionality of the TDP-43. We recently demonstrated that brain ischemia triggers an age-dependent deregulation of TDP-43 that was associated with exacerbated neurodegeneration. Here, we report that chronic cerebral hypoperfusion in mice (CCH) produced by unilateral common carotid artery occlusion induces cytoplasmic mislocalization of TDP-43 and formation of insoluble phosho-TDP-43 aggregates reminiscent of pathological changes detected in cortical neurons of human brain samples from patients suffering from vascular dementia. Moreover, the CCH in mice caused chronic activation of microglia and innate immune response, development of cognitive deficits, and motor impairments. Oral administration of a novel analog (IMS-088) of withaferin A, an antagonist of nuclear factor-κB essential modulator (NEMO), led to mitigation of TDP-43 pathology, enhancement of autophagy, and amelioration of cognitive/motor deficits in CCH mice. Taken together, our results suggest that targeting TDP-43 pathogenic inclusions may have a disease-modifying effect in dementia caused by chronic brain hypoperfusion.


Subject(s)
Cerebrovascular Circulation/drug effects , Cerebrovascular Disorders/genetics , Cognitive Dysfunction/genetics , DNA-Binding Proteins/genetics , Motor Disorders/genetics , TDP-43 Proteinopathies/genetics , Animals , Cerebrovascular Circulation/physiology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/pathology , Chronic Disease , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Drug Delivery Systems/methods , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Disorders/drug therapy , Motor Disorders/pathology , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/pathology , Withanolides/administration & dosage , Withanolides/chemistry
3.
JCI Insight ; 5(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-33021970

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), 2 incurable neurodegenerative disorders, share the same pathological hallmark named TDP43 (TAR DNA binding protein 43) proteinopathy. This event is characterized by a consistent cytoplasmic mislocalization and aggregation of the protein TDP43, which loses its physiological properties, leading neurons to death. Antibody-based approaches are now emerging interventions in the field of neurodegenerative disorders. Here, we tested the target specificity, in vivo distribution, and therapeutic efficacy of a monoclonal full-length antibody, named E6, in TDP43-related conditions. We observed that the antibody recognizes specifically the cytoplasmic fraction of TDP43. We demonstrated its ability in targeting large neurons in the spinal cord of mice and in reducing TDP43 mislocalization and NF-κB activation. We also recognized the proteasome as well as the lysosome machineries as possible mechanisms used by the antibody to reduce TDP43 proteinopathy. To our knowledge, this is the first report showing the therapeutic efficacy and feasibility of a full-length antibody against TDP43 in reducing TDP43 proteinopathy in spinal neurons of an ALS/FTLD mouse model.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Antibodies, Monoclonal/pharmacology , DNA-Binding Proteins/immunology , Neurons/drug effects , Spinal Cord/drug effects , TDP-43 Proteinopathies/drug therapy , Aged , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Case-Control Studies , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , NF-kappa B/metabolism , Neurons/immunology , Neurons/pathology , Spinal Cord/immunology , Spinal Cord/pathology , TDP-43 Proteinopathies/immunology , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology
4.
Nature ; 588(7838): 459-465, 2020 12.
Article in English | MEDLINE | ID: mdl-32866962

ABSTRACT

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Subject(s)
Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Progranulins/deficiency , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Aging/genetics , Aging/pathology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Complement Activation/drug effects , Complement Activation/immunology , Complement C1q/antagonists & inhibitors , Complement C1q/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/immunology , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Male , Mice , Nuclear Pore/metabolism , Nuclear Pore/pathology , Progranulins/genetics , RNA-Seq , Single-Cell Analysis , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Thalamus/metabolism , Thalamus/pathology , Transcriptome
5.
J Neurophysiol ; 121(1): 285-297, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30461368

ABSTRACT

Almost 90% of amyotrophic lateral sclerosis (ALS) cases are characterized by the presence of aggregates of insoluble, misfolded cytoplasmic TAR DNA binding protein of 43 kDa (TDP-43). Distal axonopathy with impaired neuromuscular junctions (NMJs) before motor neuron degeneration or clinical onset of symptoms has been hypothesized as an early pathology in ALS. However, synaptic defects at the NMJ caused by TDP-43 mutations have not been characterized. In this study, we examined a previously reported zebrafish line expressing the tardbpY220X/Y220X variant, which results in an unstable and degraded protein. These tardbp-/- larvae, however, mature normally due to the upregulated expression of an alternative splice variant of the tardbp paralog tardbp-like, or tardbpl. We generated a mutant line with a CRISPR/Cas9-mediated 5-base pair deletion encompassing the ATG start codon of tardbpl and in-crossed these with tardbp-/- mutants to obtain tardbp-/- and tardbpl-/- double mutants, herein referred to as hom/hom. We subsequently characterized morphological, coiling, locomotor, synaptic, and NMJ structural abnormalities in the hom/hom mutants and in their genotypic controls. We observed that hom/hom mutants displayed gross morphological defects, early lethality, reduced locomotor function, aberrant quantal transmission, and perturbed synapse architecture at the NMJ. We further employed pharmacological manipulations in an effort to rescue phenotypic defects and observed that tardbp+/-; tardbpl-/- (herein referred to as het/hom) mutants, but not hom/hom mutants, were sensitive to chronic treatments of BAY K 8644, an L-type calcium channel agonist. This result highlights the importance of partial vs. complete loss of allelic functions of TDP-43. NEW & NOTEWORTHY This study highlights the importance of partial vs. complete loss of allelic functions of TDP-43 in a zebrafish loss of function model, thus making it an attractive tool for drug screening approaches.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Loss of Function Mutation , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Alleles , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Genotype , Motor Activity/drug effects , Motor Activity/physiology , Neuromuscular Junction/drug effects , Neuromuscular Junction/growth & development , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Zebrafish
6.
Sci Rep ; 8(1): 16002, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375462

ABSTRACT

Findings from studies using animal models expressing amyotrophic lateral sclerosis (ALS) mutations in RNA-binding proteins, such as Transactive Response DNA-binding protein-43 (TDP-43), indicate that this protein, which is involved in multiple functions, including transcriptional regulation and pre-mRNA splicing, represents a key candidate in ALS development. This study focuses on characterizing, in a Drosophila genetic model of ALS (TDP-43), the effects of Mucuna pruriens (Mpe) and Withania somnifera (Wse). Electrophysiological and behavioural data in TDP-43 mutant flies revealed anomalous locomotion (i.e. impaired climbing with unexpected hyperactivity) and sleep dysregulation. These features, in agreement with previous findings with a different ALS model, were at least partially, rescued by treatment with Mpe and Wse. In addition, electrophysiological recordings from dorsal longitudinal muscle fibers and behavioral observations of TDP-43 flies exposed to the volatile anaesthetics, diethyl ether or chloroform, showed paradoxical responses, which were normalized upon Mpe or Wse treatment. Hence, given the involvement of some potassium channels in the effects of anaesthetics, our results also hint toward a possible dysregulation of some potassium channels in the ALS-TDP-43 Drosophila model, that might shed new light on future therapeutic strategies pertaining to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Electrophysiological Phenomena/drug effects , Locomotion/drug effects , Phytochemicals/pharmacology , Plant Extracts/pharmacology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/physiopathology , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Disease Models, Animal , Drosophila melanogaster , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Motor Neurons/drug effects , Motor Neurons/metabolism , Mutation , Phytochemicals/chemistry , Plant Extracts/chemistry , TDP-43 Proteinopathies/drug therapy
7.
Neurobiol Dis ; 118: 40-54, 2018 10.
Article in English | MEDLINE | ID: mdl-29940336

ABSTRACT

The antiepileptic drug ethosuximide has recently been shown to be neuroprotective in various Caenorhabditis elegans and rodent neurodegeneration models. It is therefore a promising repurposing candidate for the treatment of multiple neurodegenerative diseases. However, high concentrations of the drug are required for its protective effects in animal models, which may impact on its translational potential and impede the identification of its molecular mechanism of action. Therefore, we set out to develop more potent neuroprotective lead compounds based on ethosuximide as a starting scaffold. Chemoinformatic approaches were used to identify compounds with structural similarity to ethosuximide and to prioritise these based on good predicated blood-brain barrier permeability and C. elegans bioaccumulation properties. Selected compounds were initially screened for anti-convulsant activity in a C. elegans pentylenetetrazol-induced seizure assay, as a rapid primary readout of bioactivity; and then assessed for neuroprotective properties in a C. elegans TDP-43 proteinopathy model based on pan-neuronal expression of human A315T mutant TDP-43. The most potent compound screened, α-methyl-α-phenylsuccinimide (MPS), ameliorated the locomotion defects and extended the shortened lifespan of TDP-43 mutant worms. MPS also directly protected against neurodegeneration by reducing the number of neuronal breaks and cell body losses in GFP-labelled GABAergic motor neurons. Importantly, optimal neuroprotection was exhibited by external application of 50 µM MPS, compared to 8 mM for ethosuximide. This greater potency of MPS was not due to bioaccumulation to higher internal levels within the worm, based on 1H-nuclear magnetic resonance analysis. Like ethosuximide, the activity of MPS was abolished by mutation of the evolutionarily conserved FOXO transcription factor, daf-16, suggesting that both compounds act via the same neuroprotective pathway(s). In conclusion, we have revealed a novel neuroprotective activity of MPS that is >100-fold more potent than ethosuximide. This increased potency will facilitate future biochemical studies to identify the direct molecular target(s) of both compounds, as we have shown here that they share a common downstream DAF-16-dependent mechanism of action. Furthermore, MPS is the active metabolite of another approved antiepileptic drug, methsuximide. Therefore, methsuximide may have repurposing potential for treatment of TDP-43 proteinopathies and possibly other human neurodegenerative diseases.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Disease Models, Animal , Succinimides/therapeutic use , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Animals , Animals, Genetically Modified , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Caenorhabditis elegans , Female , Male , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Succinimides/chemistry , TDP-43 Proteinopathies/pathology
8.
Brain ; 140(12): 3081-3104, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29053785

ABSTRACT

Progranulin, a secreted glycoprotein, is encoded in humans by the single GRN gene. Progranulin consists of seven and a half, tandemly repeated, non-identical copies of the 12 cysteine granulin motif. Many cellular processes and diseases are associated with this unique pleiotropic factor that include, but are not limited to, embryogenesis, tumorigenesis, inflammation, wound repair, neurodegeneration and lysosome function. Haploinsufficiency caused by autosomal dominant mutations within the GRN gene leads to frontotemporal lobar degeneration, a progressive neuronal atrophy that presents in patients as frontotemporal dementia. Frontotemporal dementia is an early onset form of dementia, distinct from Alzheimer's disease. The GRN-related form of frontotemporal lobar dementia is a proteinopathy characterized by the appearance of neuronal inclusions containing ubiquitinated and fragmented TDP-43 (encoded by TARDBP). The neurotrophic and neuro-immunomodulatory properties of progranulin have recently been reported but are still not well understood. Gene delivery of GRN in experimental models of Alzheimer's- and Parkinson's-like diseases inhibits phenotype progression. Here we review what is currently known concerning the molecular function and mechanism of action of progranulin in normal physiological and pathophysiological conditions in both in vitro and in vivo models. The potential therapeutic applications of progranulin in treating neurodegenerative diseases are highlighted.


Subject(s)
Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Animals , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/drug therapy , Frontotemporal Lobar Degeneration/metabolism , Granulins , Haploinsufficiency , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Models, Neurological , Molecular Targeted Therapy , Mutation , Progranulins , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism
9.
Brain ; 139(Pt 12): 3187-3201, 2016 12.
Article in English | MEDLINE | ID: mdl-27679482

ABSTRACT

Cytoplasmic TDP-43 aggregation is a pathological hallmark of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Here we investigated the role of exosomes in the secretion and propagation of TDP-43 aggregates. TDP-43 was detected in secreted exosomes from Neuro2a cells and primary neurons but not from astrocytes or microglia. Evidence is presented that protein aggregation and autophagy inhibition are factors that promote exosomal secretion of TDP-43. We also report that levels of exosomal TDP-43 full length and C-terminal fragment species are upregulated in human amyotrophic lateral sclerosis brains. Exposure of Neuro2a cells to exosomes from amyotrophic lateral sclerosis brain, but not from control brain, caused cytoplasmic redistribution of TDP-43, suggesting that secreted exosomes might contribute to propagation of TDP-43 proteinopathy. Yet, inhibition of exosome secretion by inactivation of neutral sphingomyelinase 2 with GW4869 or by silencing RAB27A provoked formation of TDP-43 aggregates in Neuro2a cells. Moreover, administration of GW4869 exacerbated the disease phenotypes of transgenic mice expressing human TDP-43A315T mutant. Thus, even though results suggest that exosomes containing pathological TDP-43 may play a key role in the propagation of TDP-43 proteinopathy, a therapeutic strategy for amyotrophic lateral sclerosis based on inhibition of exosome production would seem inappropriate, as in vivo data suggest that exosome secretion plays an overall beneficial role in neuronal clearance of pathological TDP-43.


Subject(s)
Aniline Compounds/pharmacology , Behavior, Animal/drug effects , Benzylidene Compounds/pharmacology , DNA-Binding Proteins/metabolism , Exosomes/metabolism , Sphingomyelin Phosphodiesterase/drug effects , TDP-43 Proteinopathies/metabolism , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Transgenic , TDP-43 Proteinopathies/drug therapy
10.
J Biotechnol ; 207: 34-8, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-25987361

ABSTRACT

TAR DNA binding protein (TDP43) is a DNA- and RNA-binding protein that is implicated in several neurodegenerative disorders termed as "TDP43 proteinopathies" including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS) and fronto-temporal lobe dementia (FTLD). We have developed an InCell Western (ICW) technique for screening TDP targeting drugs in 96 well plates. We tested 281 compounds and identified a novel compound hexachlorophene (referred to as B10) that showed potent reduction in TDP43 levels. The effect of B10 on TDP protein level was validated in two different cellular models: endogenous TDP43 expressing N9 microglial cells and TDP43-over-expressing HEK293 and HeLa cells. We also analyzed effect of B10 on various pathological forms of TDP such as the C25 cleaved fragment that localizes to the cytosol, insoluble high molecular weight species, and ALS-linked mutants. Our data suggest that B10 effectively reduces all forms of TDP. Overall, our data suggest that B10 could serve as a potential drug molecule for the treatment of AD, ALS and other TDP43 proteinopathies.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drug Discovery/methods , Hexachlorophene/pharmacology , Animals , Cell Line , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Hexachlorophene/chemistry , Humans , Mice , Molecular Targeted Therapy , TDP-43 Proteinopathies/drug therapy
11.
Ther Umsch ; 72(4): 279-85, 2015 Apr.
Article in German | MEDLINE | ID: mdl-25791052

ABSTRACT

The most prevalent causes of dementia are progressive and irreversible neurodegenerative diseases of the brain. Alzheimer's disease ranks first and is follwed by Parkinson and Lewy body disease as well as the Frontotemporal lobar degenerations. These neurodegenerative processes are characterised by the production, aggregation and deposition of pathological proteins. These are ß amyloid and tau in Alzheimer's disease; α synuclein in der Parkinson's- and Lewy body disease, and tau, TDP-43 as well as FUS in the Frontotemporal lobar degenerations. Aggregation into oligomers and fibrils and subsequent sedimentation of these proteins lead to nerve cell dysfunction, synaptic failure and ultimately to the demise of neurons. The deficits and imbalance of neurotransmitter systems which represent an important target of the current pharmacological treatment of dementia are consequences of nerve cell loss. Many of the novel treatment approaches that are being tested in clinical trials are aimed at preventing, slowing or ameliorating the production, aggregation and deposition of pathological proteins. Key strategies are inhibition of secretases which generate ß amyloid, active and passive immunisation against ß amyloid, restriction ß amyloid and tau aggregation as well as stimulation of ß amyloid clearance. In addition clinical trials are ongoing on symptomatic treatments including the simultaneous stimulation of multiple neurotransmitter systems, compensation of brain insulin resistance, and neuroprotection through certain nutrients. In addition to novel drug treatments non-pharmacological interventions are also being developed.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Aged , Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Brain/physiopathology , Humans , Immunization, Passive , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/immunology , Neurotransmitter Agents/metabolism , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/etiology , TDP-43 Proteinopathies/physiopathology , Vaccination
12.
Neurobiol Aging ; 35(1): 79-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23954172

ABSTRACT

Accumulation of the transactive response DNA-binding protein 43 (TDP-43) is a major hallmark of several neurodegenerative disorders, collectively known as TDP-43 proteinopathies. The most common TDP-43 proteinopathies, frontotemporal lobar degeneration with TDP-43-positive inclusions, and amyotrophic lateral sclerosis, share overlapping neuropathological and clinical phenotypes. The development and detailed analysis of animal models of TDP-43 proteinopathies are critical for understanding the pathogenesis of these disorders. Transgenic mice overexpressing mutant human TDP-43 (herein referred to as hTDP-43) are characterized by neurodegeneration and reduced life span. However, little is known about the behavioral phenotype of these mice. Here we report the novel finding that hTDP-43 mice develop deficits in cognition, motor performance, and coordination. We show that these behavioral deficits are associated with the accumulation of nuclear and cytosolic TDP-43 C-terminal fragments, a decrease in endogenous TDP-43 levels, and synaptic loss. Our findings provide critical insights into disease pathology, and will help guide future preclinical studies aimed at testing the effects of potential therapeutic agents on the onset and progression of TDP-43 proteinopathies.


Subject(s)
Cognition Disorders/genetics , Cognition Disorders/pathology , DNA-Binding Proteins/metabolism , Peptide Fragments/metabolism , Synapses/pathology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , Amyotrophic Lateral Sclerosis , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Frontotemporal Lobar Degeneration , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Mutation , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/pathology
13.
J Neurochem ; 129(2): 350-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24298989

ABSTRACT

The E3 ubiquitin ligase Parkin plays a central role in the pathogenesis of many neurodegenerative diseases. Parkin promotes specific ubiquitination and affects the localization of transactivation response DNA-binding protein 43 (TDP-43), which controls the translation of thousands of mRNAs. Here we tested the effects of lentiviral Parkin and TDP-43 expression on amino acid metabolism in the rat motor cortex using high frequency ¹³C NMR spectroscopy. TDP-43 expression increased glutamate levels, decreased the levels of other amino acids, including glutamine, aspartate, leucine and isoleucine, and impaired mitochondrial tricarboxylic acid cycle. TDP-43 induced lactate accumulation and altered the balance between excitatory (glutamate) and inhibitory (GABA) neurotransmitters. Parkin restored amino acid levels, neurotransmitter balance and tricarboxylic acid cycle metabolism, rescuing neurons from TDP-43-induced apoptotic death. Furthermore, TDP-43 expression led to an increase in 4E-BP levels, perhaps altering translational control and deregulating amino acid synthesis; while Parkin reversed the effects of TDP-43 on the 4E-BP signaling pathway. Taken together, these data suggest that Parkin may affect TDP-43 localization and mitigate its effects on 4E-BP signaling and loss of amino acid homeostasis.


Subject(s)
Amino Acids/metabolism , Cell Death/drug effects , TDP-43 Proteinopathies/drug therapy , Ubiquitin-Protein Ligases/pharmacology , Animals , Blotting, Western , Carrier Proteins/metabolism , Caspase 3/metabolism , Citric Acid Cycle/drug effects , Fluorometry , Genetic Vectors , Homeostasis/drug effects , Homeostasis/physiology , Intracellular Signaling Peptides and Proteins , Lentivirus/genetics , Magnetic Resonance Spectroscopy , Male , Motor Cortex/drug effects , Motor Cortex/metabolism , Neurotransmitter Agents/metabolism , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , TDP-43 Proteinopathies/pathology , TOR Serine-Threonine Kinases/metabolism , gamma-Aminobutyric Acid/metabolism
14.
J Biol Chem ; 288(27): 19614-24, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23689371

ABSTRACT

TDP-43 (TAR DNA-binding protein of 43 kDa) is a major deposited protein in amyotrophic lateral sclerosis and frontotemporal dementia with ubiquitin. A great number of genetic mutations identified in the flexible C-terminal region are associated with disease pathologies. We investigated the molecular determinants of TDP-43 aggregation and its underlying mechanisms. We identified a hydrophobic patch (residues 318-343) as the amyloidogenic core essential for TDP-43 aggregation. Biophysical studies demonstrated that the homologous peptide formed a helix-turn-helix structure in solution, whereas it underwent structural transformation from an α-helix to a ß-sheet during aggregation. Mutation or deletion of this core region significantly reduced the aggregation and cytoplasmic inclusions of full-length TDP-43 (or TDP-35 fragment) in cells. Thus, structural transformation of the amyloidogenic core initiates the aggregation and cytoplasmic inclusion formation of TDP-43. This particular core region provides a potential therapeutic target to design small-molecule compounds for mitigating TDP-43 proteinopathies.


Subject(s)
Amyloid/metabolism , DNA-Binding Proteins/metabolism , Inclusion Bodies/metabolism , Amyloid/genetics , Animals , Caenorhabditis elegans , DNA-Binding Proteins/genetics , Drug Design , HeLa Cells , Helix-Turn-Helix Motifs , Humans , Hydrophobic and Hydrophilic Interactions , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Protein Structure, Tertiary , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology
15.
Ann Neurol ; 74(1): 39-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23424178

ABSTRACT

OBJECTIVE: Kinase hyperactivity occurs in both neurodegenerative disease and cancer. Lesions containing hyperphosphorylated aggregated TDP-43 characterize amyotrophic lateral sclerosis and frontotemporal lobar degeneration with TDP-43 inclusions. Dual phosphorylation of TDP-43 at serines 409/410 (S409/410) drives neurotoxicity in disease models; therefore, TDP-43-specific kinases are candidate targets for intervention. METHODS: To find therapeutic targets for the prevention of TDP-43 phosphorylation, we assembled and screened a comprehensive RNA interference library targeting kinases in TDP-43 transgenic Caenorhabditis elegans. RESULTS: We show CDC7 robustly phosphorylates TDP-43 at pathological residues S409/410 in C. elegans, in vitro, and in human cell culture. In frontotemporal lobar degeneration (FTLD)-TDP cases, CDC7 immunostaining overlaps with the phospho-TDP-43 pathology found in frontal cortex. Furthermore, PHA767491, a small molecule inhibitor of CDC7, reduces TDP-43 phosphorylation and prevents TDP-43-dependent neurodegeneration in TDP-43-transgenic animals. INTERPRETATION: Taken together, these data support CDC7 as a novel therapeutic target for TDP-43 proteinopathies, including FTLD-TDP and amyotrophic lateral sclerosis.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neurodegenerative Diseases/etiology , Protein Serine-Threonine Kinases/metabolism , TDP-43 Proteinopathies/therapy , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cell Line, Transformed , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Movement/physiology , Mutation/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Phosphorylation , Piperidones/pharmacology , Pyrroles/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serine/metabolism , TDP-43 Proteinopathies/complications , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Transfection
16.
J Neurosci ; 33(3): 906-13, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325230

ABSTRACT

The accumulation of TDP-43 (transactive response DNA-binding protein 43) and its 25 kDa C-terminal fragment (TDP-25) is a hallmark of several neurodegenerative disorders, including frontotemporal lobar degeneration (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). The majority of FTLD-TDP cases are due to loss of function mutations in the gene encoding progranulin, a secreted growth factor. In ALS, specific mutations in the gene encoding TDP-43 have been linked to the disease pathogenesis. In both cases, however, the penetrance of the mutations greatly increases during aging, suggesting that other genetic or environmental factors may facilitate the development of the disease. Using transgenic mice that overexpress the 25 kDa C-terminal fragment of TDP-43, here we show that glucocorticoids, stress hormones known to increase the brain susceptibility to neurotoxic insults, increase the levels of soluble TDP-25 and exacerbate cognitive deficits, without altering full-length TDP-43 levels. Additionally, we show that the mechanism underlying the glucocorticoid-mediated increase in TDP-25 levels is coupled to changes in the glutathione redox state. Glutathione is an antioxidant involved in protecting cells from damage caused by reactive oxygen species; notably, alterations in the ratio of reduced to oxidized glutathione, which is the primary determinant of the cellular redox state, are associated with aging and neurodegeneration. We show that restoring the ratio of reduced to oxidized glutathione blocks the glucocorticoid effects on TDP-25. These data show that glucocorticoids potentiate the neurotoxic action of TDP-25 by increasing its levels and clearly indicate the role of cellular oxidative damage in this process.


Subject(s)
Aging/metabolism , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Glutathione/metabolism , Maze Learning/drug effects , TDP-43 Proteinopathies/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , DNA-Binding Proteins/genetics , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Glutathione Disulfide/metabolism , Maze Learning/physiology , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology
17.
Proc Natl Acad Sci U S A ; 109(37): 15024-9, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22932872

ABSTRACT

TDP-43 is a multifunctional DNA/RNA-binding protein that has been identified as the major component of the cytoplasmic ubiquitin (+) inclusions (UBIs) in diseased cells of frontotemporal lobar dementia (FTLD-U) and amyotrophic lateral sclerosis (ALS). Unfortunately, effective drugs for these neurodegenerative diseases are yet to be developed. We have tested the therapeutic potential of rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR) and three other autophagy activators (spermidine, carbamazepine, and tamoxifen) in a FTLD-U mouse model with TDP-43 proteinopathies. Rapamycin treatment has been reported to be beneficial in some animal models of neurodegenerative diseases but not others. Furthermore, the effects of rapamycin treatment in FTLD-U have not been investigated. We show that rapamycin treatment effectively rescues the learning/memory impairment of these mice at 3 mo of age, and it significantly slows down the age-dependent loss of their motor function. These behavioral improvements upon rapamycin treatment are accompanied by a decreased level of caspase-3 and a reduction of neuron loss in the forebrain of FTLD-U mice. Furthermore, the number of cells with cytosolic TDP-43 (+) inclusions and the amounts of full-length TDP-43 as well as its cleavage products (35 kDa and 25 kDa) in the urea-soluble fraction of the cellular extract are significantly decreased upon rapamycin treatment. These changes in TDP-43 metabolism are accompanied by rapamycin-induced decreases in mTOR-regulated phospho-p70 S6 kinase (P-p70) and the p62 protein, as well as increases in the autophagic marker LC3. Finally, rapamycin as well as spermidine, carbamazepine, and tamoxifen could also rescue the motor dysfunction of 7-mo-old FTLD-U mice. These data suggest that autophagy activation is a potentially useful route for the therapy of neurodegenerative diseases with TDP-43 proteinopathies.


Subject(s)
Autophagy/drug effects , Frontotemporal Dementia/complications , Frontotemporal Dementia/drug therapy , Sirolimus/pharmacology , TDP-43 Proteinopathies/complications , TDP-43 Proteinopathies/drug therapy , Analysis of Variance , Animals , Blotting, Western , Carbamazepine/pharmacology , Caspase 3/metabolism , Fluorometry , Frontotemporal Dementia/physiopathology , In Situ Nick-End Labeling , Maze Learning/drug effects , Memory Disorders/drug therapy , Mice , Psychomotor Performance/drug effects , Rotarod Performance Test , Sirolimus/therapeutic use , Spermidine/pharmacology , TDP-43 Proteinopathies/physiopathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tamoxifen/pharmacology
18.
J Clin Invest ; 122(1): 107-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22156203

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron degeneration, which ultimately leads to paralysis and death. Mutation of TAR DNA binding protein 43 (TDP-43) has been linked to the development of an inherited form of ALS. Existing TDP-43 transgenic animals develop a limited loss of motor neurons and therefore do not faithfully reproduce the core phenotype of ALS. Here, we report the creation of multiple lines of transgenic rats in which expression of ALS-associated mutant human TDP-43 is restricted to either motor neurons or other types of neurons and skeletal muscle and can be switched on and off. All of these rats developed progressive paralysis reminiscent of ALS when the transgene was switched on. Rats expressing mutant TDP-43 in motor neurons alone lost more spinal motor neurons than rats expressing the disease gene in varying neurons and muscle cells, although these rats all developed remarkable denervation atrophy of skeletal muscles. Intriguingly, progression of the disease was halted after transgene expression was switched off; in rats with limited loss of motor neurons, we observed a dramatic recovery of motor function, but in rats with profound loss of motor neurons, we only observed a moderate recovery of motor function. Our finding suggests that mutant TDP-43 in motor neurons is sufficient to promote the onset and progression of ALS and that motor neuron degeneration is partially reversible, at least in mutant TDP-43 transgenic rats.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Motor Neurons/physiology , Mutant Proteins/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Base Sequence , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Doxycycline/pharmacology , Humans , Motor Neurons/drug effects , Motor Neurons/pathology , Mutant Proteins/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Rats , Rats, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology , TDP-43 Proteinopathies/physiopathology
19.
J Biol Chem ; 287(6): 4107-20, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22147697

ABSTRACT

No current therapies target the underlying cellular pathologies of age-related neurodegenerative diseases. Model organisms provide a platform for discovering compounds that protect against the toxic, misfolded proteins that initiate these diseases. One such protein, TDP-43, is implicated in multiple neurodegenerative diseases, including amyotrophic lateral sclerosis and frontotemporal lobar degeneration. In yeast, TDP-43 expression is toxic, and genetic modifiers first discovered in yeast have proven to modulate TDP-43 toxicity in both neurons and humans. Here, we describe a phenotypic screen for small molecules that reverse TDP-43 toxicity in yeast. One group of hit compounds was 8-hydroxyquinolines (8-OHQ), a class of clinically relevant bioactive metal chelators related to clioquinol. Surprisingly, in otherwise wild-type yeast cells, different 8-OHQs had selectivity for rescuing the distinct toxicities caused by the expression of TDP-43, α-synuclein, or polyglutamine proteins. In fact, each 8-OHQ synergized with the other, clearly establishing that they function in different ways. Comparative growth and molecular analyses also revealed that 8-OHQs have distinct metal chelation and ionophore activities. The diverse bioactivity of 8-OHQs indicates that altering different aspects of metal homeostasis and/or metalloprotein activity elicits distinct protective mechanisms against several neurotoxic proteins. Indeed, phase II clinical trials of an 8-OHQ has produced encouraging results in modifying Alzheimer disease. Our unbiased identification of 8-OHQs in a yeast TDP-43 toxicity model suggests that tailoring 8-OHQ activity to a particular neurodegenerative disease may be a viable therapeutic strategy.


Subject(s)
DNA-Binding Proteins/metabolism , Models, Biological , Oxyquinoline/pharmacology , Polyglutamic Acid/metabolism , TDP-43 Proteinopathies/drug therapy , alpha-Synuclein/metabolism , Animals , Caenorhabditis elegans , Chelating Agents/pharmacology , DNA-Binding Proteins/genetics , Humans , Ionophores/pharmacology , Polyglutamic Acid/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , alpha-Synuclein/genetics
20.
Rinsho Shinkeigaku ; 49(11): 783-5, 2009 Nov.
Article in Japanese | MEDLINE | ID: mdl-20030209

ABSTRACT

The TDP-43 proteinopathies: Toward understanding of the molecular pathogenesis. TAR DNA binding protein of 43 kDa (TDP-43), a heterogeneous nuclear ribonucleoprotein was identified as a major component of ubiquitin-positive inclusions in FTLD and ALS, and the concept of TDP-43 proteinopathies was proposed. Immunoblot and immunohistochemical analyses using multiple anti-phosphorylated TDP-43 antibodies revealed that hyperphosphorylated 18-26 kDa C-terminal fragments in addition to the full-length TDP-43 are major constituents of inclusions in FTLD-U and ALS. Recent discovery of mutations in the TDP-43 gene in familial and sporadic ALS, indicating that abnormality of TDP-43 protein cause neurodegeneration. It also strongly suggests that aggregation of TDP-43 or the process is responsible for neurodegeneration in FTLD-U and ALS. To investigate the molecular mechanisms of aggregation of TDP-43, we have established two cellular models for intracellular aggregates of TDP-43 similar to those in brains of TDP-43 proteinopathies patients. The first consists of SH-SY5Y cells expressing mutant TDP-43 that lacks both the nuclear localization signal (NLS) and residues 187-192 (deltaNLS & 187-192). The second model consists of SH-SY5Y cells expressing an aggregation-prone TDP-43 C-terminal fragment as a green fluorescent protein (GFP)-fusion. In these cells, round structures positive for both anti-pS409/410 and anti-Ub are observed. These results suggest that intracellular localization of TDP-43, truncation of TDP-43 and proteasomal dysfunction of cells may be involved in the pathological process of TDP-43 proteinopathies. We also found that two small compounds that have been reported to be beneficial in phase II clinical trials of Alzheimer's disease, inhibited the formation of TDP-43 aggregates in these two cellular models, suggesting that these compounds may be effective for the treatment of ALS and FTLD-U.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/metabolism , TDP-43 Proteinopathies/etiology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/genetics , Clinical Trials, Phase II as Topic , DNA-Binding Proteins/genetics , Drug Design , Humans , Mutation , Nuclear Localization Signals , Phosphorylation , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Ubiquitin
SELECTION OF CITATIONS
SEARCH DETAIL
...