Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 40(8): e103811, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33644875

RESUMO

HSP27 is a human molecular chaperone that forms large, dynamic oligomers and functions in many aspects of cellular homeostasis. Mutations in HSP27 cause Charcot-Marie-Tooth (CMT) disease, the most common inherited disorder of the peripheral nervous system. A particularly severe form of CMT disease is triggered by the P182L mutation in the highly conserved IxI/V motif of the disordered C-terminal region, which interacts weakly with the structured core domain of HSP27. Here, we observed that the P182L mutation disrupts the chaperone activity and significantly increases the size of HSP27 oligomers formed in vivo, including in motor neurons differentiated from CMT patient-derived stem cells. Using NMR spectroscopy, we determined that the P182L mutation decreases the affinity of the HSP27 IxI/V motif for its own core domain, leaving this binding site more accessible for other IxI/V-containing proteins. We identified multiple IxI/V-bearing proteins that bind with higher affinity to the P182L variant due to the increased availability of the IxI/V-binding site. Our results provide a mechanistic basis for the impact of the P182L mutation on HSP27 and suggest that the IxI/V motif plays an important, regulatory role in modulating protein-protein interactions.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Choque Térmico/química , Chaperonas Moleculares/química , Adulto , Sítios de Ligação , Células Cultivadas , Células HeLa , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Simulação de Dinâmica Molecular , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica , Multimerização Proteica
2.
Brain ; 146(7): 2885-2896, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-36511878

RESUMO

Charcot-Marie-Tooth disease is the most common inherited disorder of the PNS. CMT1A accounts for 40-50% of all cases and is caused by a duplication of the PMP22 gene on chromosome 17, leading to dysmyelination in the PNS. Patient-derived models to study such myelination defects are lacking as the in vitro generation of human myelinating Schwann cells has proved to be particularly challenging. Here, we present an induced pluripotent stem cell-derived organoid culture, containing various cell types of the PNS, including myelinating human Schwann cells, which mimics the human PNS. Single-cell analysis confirmed the PNS-like cellular composition and provides insight into the developmental trajectory. We used this organoid model to study disease signatures of CMT1A, revealing early ultrastructural myelin alterations, including increased myelin periodic line distance and hypermyelination of small axons. Furthermore, we observed the presence of onion-bulb-like formations in a later developmental stage. These hallmarks were not present in the CMT1A-corrected isogenic line or in a CMT2A iPSC line, supporting the notion that these alterations are specific to CMT1A. Downregulation of PMP22 expression using short-hairpin RNAs or a combinatorial drug consisting of baclofen, naltrexone hydrochloride and D-sorbitol was able to ameliorate the myelin defects in CMT1A-organoids. In summary, this self-organizing organoid model can capture biologically meaningful features of the disease and capture the physiological complexity, forms an excellent model for studying demyelinating diseases and supports the therapeutic approach of reducing PMP22 expression.


Assuntos
Doença de Charcot-Marie-Tooth , Células-Tronco Pluripotentes Induzidas , Humanos , Bainha de Mielina/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Regulação para Baixo , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Organoides/metabolismo , Células de Schwann
3.
Brain ; 144(8): 2471-2485, 2021 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-34128983

RESUMO

Axonal Charcot-Marie-Tooth neuropathies (CMT type 2) are caused by inherited mutations in various genes functioning in different pathways. The types of genes and multiplicity of mutations reflect the clinical and genetic heterogeneity in CMT2 disease, which complicates its diagnosis and has inhibited the development of therapies. Here, we used CMT2 patient-derived pluripotent stem cells (iPSCs) to identify common hallmarks of axonal degeneration shared by different CMT2 subtypes. We compared the cellular phenotypes of neurons differentiated from CMT2 patient iPSCs with those from healthy controls and a CRISPR/Cas9-corrected isogenic line. Our results demonstrated neurite network alterations along with extracellular electrophysiological abnormalities in the differentiated motor neurons. Progressive deficits in mitochondrial and lysosomal trafficking, as well as in mitochondrial morphology, were observed in all CMT2 patient lines. Differentiation of the same CMT2 iPSC lines into peripheral sensory neurons only gave rise to cellular phenotypes in subtypes with sensory involvement, supporting the notion that some gene mutations predominantly affect motor neurons. We revealed a common mitochondrial dysfunction in CMT2-derived motor neurons, supported by alterations in the expression pattern and oxidative phosphorylation, which could be recapitulated in the sciatic nerve tissue of a symptomatic mouse model. Inhibition of a dual leucine zipper kinase could partially ameliorate the mitochondrial disease phenotypes in CMT2 subtypes. Altogether, our data reveal shared cellular phenotypes across different CMT2 subtypes and suggests that targeting such common pathomechanisms could allow the development of a uniform treatment for CMT2.


Assuntos
Doença de Charcot-Marie-Tooth/metabolismo , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Mitocôndrias/patologia , Neurônios Motores/patologia , Mutação , Linhagem
4.
Acta Neuropathol ; 135(1): 131-148, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28780615

RESUMO

Mutations in the small heat shock protein B8 gene (HSPB8/HSP22) have been associated with distal hereditary motor neuropathy, Charcot-Marie-Tooth disease, and recently distal myopathy. It is so far not clear how mutant HSPB8 induces the neuronal and muscular phenotypes and if a common pathogenesis lies behind these diseases. Growing evidence points towards a role of HSPB8 in chaperone-associated autophagy, which has been shown to be a determinant for the clearance of poly-glutamine aggregates in neurodegenerative diseases but also for the maintenance of skeletal muscle myofibrils. To test this hypothesis and better dissect the pathomechanism of mutant HSPB8, we generated a new transgenic mouse model leading to the expression of the mutant protein (knock-in lines) or the loss-of-function (functional knock-out lines) of the endogenous protein Hspb8. While the homozygous knock-in mice developed motor deficits associated with degeneration of peripheral nerves and severe muscle atrophy corroborating patient data, homozygous knock-out mice had locomotor performances equivalent to those of wild-type animals. The distal skeletal muscles of the post-symptomatic homozygous knock-in displayed Z-disk disorganisation, granulofilamentous material accumulation along with Hspb8, αB-crystallin (HSPB5/CRYAB), and desmin aggregates. The presence of the aggregates correlated with reduced markers of effective autophagy. The sciatic nerve of the homozygous knock-in mice was characterized by low autophagy potential in pre-symptomatic and Hspb8 aggregates in post-symptomatic animals. On the other hand, the sciatic nerve of the homozygous knock-out mice presented a normal morphology and their distal muscle displayed accumulation of abnormal mitochondria but intact myofiber and Z-line organisation. Our data, therefore, suggest that toxic gain-of-function of mutant Hspb8 aggregates is a major contributor to the peripheral neuropathy and the myopathy. In addition, mutant Hspb8 induces impairments in autophagy that may aggravate the phenotype.


Assuntos
Miopatias Distais/metabolismo , Mutação com Ganho de Função , Proteínas de Choque Térmico HSP20/genética , Proteínas de Choque Térmico HSP20/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miopatias Congênitas Estruturais/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Atrofia/metabolismo , Atrofia/patologia , Autofagia/fisiologia , Modelos Animais de Doenças , Miopatias Distais/patologia , Feminino , Proteínas de Choque Térmico , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Chaperonas Moleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miopatias Congênitas Estruturais/patologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
5.
Acta Neuropathol ; 131(4): 505-23, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26744348

RESUMO

There is a growing appreciation that membrane-bound organelles in eukaryotic cells communicate directly with one another through direct membrane contact sites. Mitochondria-associated membranes are specialized subdomains of the endoplasmic reticulum that function as membrane contact sites between the endoplasmic reticulum and mitochondria. These sites have emerged as major players in lipid metabolism and calcium signaling. More recently also autophagy and mitochondrial dynamics have been found to be regulated at ER-mitochondria contact sites. Neurons critically depend on mitochondria-associated membranes as a means to exchange metabolites and signaling molecules between these organelles. This is underscored by the fact that genes affecting mitochondrial and endoplasmic reticulum homeostasis are clearly overrepresented in several hereditary neurodegenerative disorders. Conversely, the processes affected by the contact sites between the endoplasmic reticulum and mitochondria are widely implicated in neurodegeneration. This review will focus on the most recent data addressing the structural composition and function of the mitochondria-associated membranes. In addition, the 3D morphology of the contact sites as observed using volume electron microscopy is discussed. Finally, it will highlight the role of several key proteins associated with these contact sites that are involved not only in dementias, amyotrophic lateral sclerosis and Parkinson's disease, but also in axonopathies such as hereditary spastic paraplegia and Charcot-Marie-Tooth disease.


Assuntos
Retículo Endoplasmático/metabolismo , Membranas Mitocondriais/metabolismo , Degeneração Neural/patologia , Neurônios/ultraestrutura , Animais , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Humanos , Degeneração Neural/metabolismo
6.
Brain ; 138(Pt 11): 3238-50, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26384929

RESUMO

The epileptic encephalopathies are a clinically and aetiologically heterogeneous subgroup of epilepsy syndromes. Most epileptic encephalopathies have a genetic cause and patients are often found to carry a heterozygous de novo mutation in one of the genes associated with the disease entity. Occasionally recessive mutations are identified: a recent publication described a distinct neonatal epileptic encephalopathy (MIM 615905) caused by autosomal recessive mutations in the SLC13A5 gene. Here, we report eight additional patients belonging to four different families with autosomal recessive mutations in SLC13A5. SLC13A5 encodes a high affinity sodium-dependent citrate transporter, which is expressed in the brain. Neurons are considered incapable of de novo synthesis of tricarboxylic acid cycle intermediates; therefore they rely on the uptake of intermediates, such as citrate, to maintain their energy status and neurotransmitter production. The effect of all seven identified mutations (two premature stops and five amino acid substitutions) was studied in vitro, using immunocytochemistry, selective western blot and mass spectrometry. We hereby demonstrate that cells expressing mutant sodium-dependent citrate transporter have a complete loss of citrate uptake due to various cellular loss-of-function mechanisms. In addition, we provide independent proof of the involvement of autosomal recessive SLC13A5 mutations in the development of neonatal epileptic encephalopathies, and highlight teeth hypoplasia as a possible indicator for SLC13A5 screening. All three patients who tried the ketogenic diet responded well to this treatment, and future studies will allow us to ascertain whether this is a recurrent feature in this severe disorder.


Assuntos
Anodontia/genética , Ácido Cítrico/metabolismo , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Simportadores/genética , Adolescente , Encefalopatias/genética , Criança , Feminino , Genes Recessivos , Predisposição Genética para Doença , Células HEK293 , Humanos , Masculino , Mutação , Linhagem , Simportadores/metabolismo
7.
Neurobiol Dis ; 65: 211-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24521780

RESUMO

Charcot-Marie-Tooth disease type 2B (CMT2B) is an inherited axonal peripheral neuropathy. It is characterised by prominent sensory loss, often complicated by severe ulcero-mutilations of toes or feet, and variable motor involvement. Missense mutations in RAB7A, the gene encoding the small GTPase Rab7, cause CMT2B and increase Rab7 activity. Rab7 is ubiquitously expressed and is involved in degradation through the lysosomal pathway. In the neurons, Rab7 plays a role in the long-range retrograde transport of signalling endosomes in the axons. Here we developed the first animal model of CMT2B, modelling one of the mutations (L129F) in Drosophila melanogaster. Behavioural assays show that this model recapitulates several hallmarks of the human disease. Upon expression of mutant Rab7 in the sensory neurons, larvae present with a reduction of temperature and pain perception. Furthermore, the larvae exhibit a crawling defect when the mutant protein is expressed in the motor neurons. Analysis of axonal transport of Rab7 positive vesicles in sensory neurons of Drosophila larvae and in neurites of mammalian neuroblastoma cells demonstrates that mutant vesicles pause less than their wild-type counterparts. This latter finding indicates that alterations in vesicle transport might contribute to the pathomechanism of CMT2B.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/fisiopatologia , Mutação/genética , Proteínas rab de Ligação ao GTP/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal/fisiologia , Diferenciação Celular , Linhagem Celular Transformada , Dendritos/patologia , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Laminopatias , Larva , Masculino , Atividade Motora/genética , Neuroblastoma/patologia , Células Receptoras Sensoriais/patologia , Transdução Genética , proteínas de unión al GTP Rab7
8.
Neurobiol Dis ; 68: 180-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24807208

RESUMO

Aminoacyl-tRNA synthetases are ubiquitously expressed proteins that charge tRNAs with their cognate amino acids. By ensuring the fidelity of protein synthesis, these enzymes are essential for the viability of every cell. Yet, mutations in six tRNA synthetases specifically affect the peripheral nerves and cause Charcot-Marie-Tooth (CMT) disease. The CMT-causing mutations in tyrosyl- and glycyl-tRNA synthetases (YARS and GARS, respectively) alter the activity of the proteins in a range of ways (some mutations do not impact charging function, while others abrogate it), making a loss of function in tRNA charging unlikely to be the cause of disease pathology. It is currently unknown which cellular mechanisms are triggered by the mutant enzymes and how this leads to neurodegeneration. Here, by expressing two pathogenic mutations (G240R, P234KY) in Drosophila, we generated a model for GARS-associated neuropathy. We observed compromised viability, and behavioral, electrophysiological and morphological impairment in flies expressing the cytoplasmic isoform of mutant GARS. Their features recapitulated several hallmarks of CMT pathophysiology and were similar to the phenotypes identified in our previously described Drosophila model of YARS-associated neuropathy. Furthermore, CG8316 and CG15599 - genes identified in a retinal degeneration screen to modify mutant YARS, also modified the mutant GARS phenotypes. Our study presents genetic evidence for common mutant-specific interactions between two CMT-associated aminoacyl-tRNA synthetases, lending support for a shared mechanism responsible for the synthetase-induced peripheral neuropathies.


Assuntos
Doença de Charcot-Marie-Tooth/complicações , Doença de Charcot-Marie-Tooth/genética , Glicina-tRNA Ligase/genética , Mutação/genética , Doenças do Sistema Nervoso Periférico/etiologia , Tirosina-tRNA Ligase/genética , Animais , Animais Geneticamente Modificados , Doença de Charcot-Marie-Tooth/patologia , Dextranos , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Humanos , Masculino , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Fibras Nervosas/fisiologia , Neurônios/patologia , Neurônios/fisiologia , Doenças do Sistema Nervoso Periférico/genética , Retina/patologia , Retina/ultraestrutura , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/etiologia , Degeneração Retiniana/genética , Rodaminas , Asas de Animais/patologia , Asas de Animais/ultraestrutura
9.
Acta Neuropathol ; 126(1): 93-108, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23728742

RESUMO

Mutations in the small heat shock protein HSPB1 (HSP27) are a cause of axonal Charcot-Marie-Tooth neuropathy (CMT2F) and distal hereditary motor neuropathy. To better understand the effect of mutations in HSPB1 on the neuronal cytoskeleton, we stably transduced neuronal cells with wild-type and mutant HSPB1 and investigated axonal transport of neurofilaments (NFs). We observed that mutant HSPB1 affected the binding of NFs to the anterograde motor protein kinesin, reducing anterograde transport of NFs. These deficits were associated with an increased phosphorylation of NFs and cyclin-dependent kinase Cdk5. As Cdk5 mediates NF phosphorylation, inhibition of Cdk5/p35 restored NF phosphorylation level, as well as NF binding to kinesin in mutant HSPB1 neuronal cells. Altogether, we demonstrate that HSPB1 mutations induce hyperphosphorylation of NFs through Cdk5 and reduce anterograde transport of NFs.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas de Choque Térmico HSP27/genética , Mutação/genética , Proteínas de Neurofilamentos/metabolismo , Transporte Axonal/genética , Axônios/metabolismo , Axônios/patologia , Linhagem Celular Tumoral , Quinase 5 Dependente de Ciclina/genética , Proteínas de Choque Térmico , Humanos , Imunoprecipitação , Cinesinas/metabolismo , Chaperonas Moleculares , Neuroblastoma/patologia , Fosforilação/genética , Transfecção/métodos
10.
Nat Cell Biol ; 25(3): 467-480, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36690850

RESUMO

Mitochondria are complex organelles with different compartments, each harbouring their own protein quality control factors. While chaperones of the mitochondrial matrix are well characterized, it is poorly understood which chaperones protect the mitochondrial intermembrane space. Here we show that cytosolic small heat shock proteins are imported under basal conditions into the mitochondrial intermembrane space, where they operate as molecular chaperones. Protein misfolding in the mitochondrial intermembrane space leads to increased recruitment of small heat shock proteins. Depletion of small heat shock proteins leads to mitochondrial swelling and reduced respiration, while aggregation of aggregation-prone substrates is countered in their presence. Charcot-Marie-Tooth disease-causing mutations disturb the mitochondrial function of HSPB1, potentially linking previously observed mitochondrial dysfunction in Charcot-Marie-Tooth type 2F to its role in the mitochondrial intermembrane space. Our results reveal that small heat shock proteins form a chaperone system that operates in the mitochondrial intermembrane space.


Assuntos
Proteínas de Choque Térmico Pequenas , Proteínas de Choque Térmico Pequenas/genética , Proteínas de Choque Térmico Pequenas/metabolismo , Chaperonas Moleculares/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo
11.
Autophagy ; 19(8): 2217-2239, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36854646

RESUMO

Chaperone-assisted selective autophagy (CASA) is a highly selective pathway for the disposal of misfolding and aggregating proteins. In muscle, CASA assures muscle integrity by favoring the turnover of structural components damaged by mechanical strain. In neurons, CASA promotes the removal of aggregating substrates. A crucial player of CASA is HSPB8 (heat shock protein family B (small) member 8), which acts in a complex with HSPA, their cochaperone BAG3, and the E3 ubiquitin ligase STUB1. Recently, four novel HSPB8 frameshift (fs) gene mutations have been linked to neuromyopathies, and encode carboxy-terminally mutated HSPB8, sharing a common C-terminal extension. Here, we analyzed the biochemical and functional alterations associated with the HSPB8_fs mutant proteins. We demonstrated that HSPB8_fs mutants are highly insoluble and tend to form proteinaceous aggregates in the cytoplasm. Notably, all HSPB8 frameshift mutants retain their ability to interact with CASA members but sequester them into the HSPB8-positive aggregates together with two autophagy receptors SQSTM1/p62 and TAX1BP1. This copartitioning process negatively affects the CASA capability to remove its clients and causes a general failure in proteostasis response. Further analyses revealed that the aggregation of the HSPB8_fs mutants occurs independently of the other CASA members or from the autophagy receptors interaction, but it is an intrinsic feature of the mutated amino acid sequence. HSPB8_fs mutants aggregation alters the differentiation capacity of muscle cells and impairs sarcomere organization. Collectively, these results shed light on a potential pathogenic mechanism shared by the HSPB8_fs mutants described in neuromuscular diseases.Abbreviations : ACD: α-crystallin domain; ACTN: actinin alpha; BAG3: BAG cochaperone 3; C: carboxy; CASA: chaperone-assisted selective autophagy; CE: carboxy-terminal extension; CLEM: correlative light and electron microscopy; CMT2L: Charcot-Marie-Tooth type 2L; CTR: carboxy-terminal region; dHMNII: distal hereditary motor neuropathy type II; EV: empty vector; FRA: filter retardation assay; fs: frameshift; HSPA/HSP70: heat shock protein family A (Hsp70); HSPB1/Hsp27: heat shock protein family B (small) member 1; HSPB8/Hsp22: heat shock protein family B (small) member 8; HTT: huntingtin; KO: knockout; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MD: molecular dynamics; MTOC: microtubule organizing center; MYH: myosin heavy chain; MYOG: myogenin; NBR1: NBR1 autophagy cargo receptor; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; NSC34: Neuroblastoma X Spinal Cord 34; OPTN: optineurin; polyQ: polyglutamine; SQSTM1/p62: sequestosome 1; STUB1/CHIP: STIP1 homology and U-box containing protein 1; TARDBP/TDP-43: TAR DNA binding protein; TAX1BP1: Tax1 binding protein 1; TUBA: tubulin alpha; WT: wild-type.


Assuntos
Doença de Charcot-Marie-Tooth , Doenças Neuromusculares , Humanos , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , Autofagia/genética , Proteínas de Choque Térmico/metabolismo , Doença de Charcot-Marie-Tooth/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo
12.
Nat Commun ; 14(1): 999, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36890170

RESUMO

Dominant mutations in tyrosyl-tRNA synthetase (YARS1) and six other tRNA ligases cause Charcot-Marie-Tooth peripheral neuropathy (CMT). Loss of aminoacylation is not required for their pathogenicity, suggesting a gain-of-function disease mechanism. By an unbiased genetic screen in Drosophila, we link YARS1 dysfunction to actin cytoskeleton organization. Biochemical studies uncover yet unknown actin-bundling property of YARS1 to be enhanced by a CMT mutation, leading to actin disorganization in the Drosophila nervous system, human SH-SY5Y neuroblastoma cells, and patient-derived fibroblasts. Genetic modulation of F-actin organization improves hallmark electrophysiological and morphological features in neurons of flies expressing CMT-causing YARS1 mutations. Similar beneficial effects are observed in flies expressing a neuropathy-causing glycyl-tRNA synthetase. Hence, in this work, we show that YARS1 is an evolutionary-conserved F-actin organizer which links the actin cytoskeleton to tRNA-synthetase-induced neurodegeneration.


Assuntos
Actinas , Tirosina-tRNA Ligase , Animais , Humanos , Actinas/metabolismo , Doença de Charcot-Marie-Tooth/genética , Drosophila/genética , Glicina-tRNA Ligase/genética , Mutação , RNA de Transferência , Tirosina-tRNA Ligase/genética , Tirosina-tRNA Ligase/metabolismo , Linhagem Celular Tumoral
13.
J Neurosci ; 31(43): 15320-8, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22031878

RESUMO

Mutations in the small heat shock protein HSPB1 (HSP27) are causative for Charcot-Marie-Tooth (CMT) neuropathy. We previously showed that a subset of these mutations displays higher chaperone activity and enhanced affinity to client proteins. We hypothesized that this excessive binding property might cause the HSPB1 mutant proteins to disturb the function of proteins essential for the maintenance or survival of peripheral neurons. In the present work, we explored this hypothesis further and compared the protein complexes formed by wild-type and mutant HSPB1. Tubulin came out as the most striking differential interacting protein, with hyperactive mutants binding more strongly to both tubulin and microtubules. This anomalous binding leads to a stabilization of the microtubule network in a microtubule-associated protein-like manner as reflected by resistance to cold depolymerization, faster network recovery after nocodazole treatment, and decreased rescue and catastrophe rates of individual microtubules. In a transgenic mouse model for mutant HSPB1 that recapitulates all features of CMT, we could confirm the enhanced interaction of mutant HSPB1 with tubulin. Increased stability of the microtubule network was also clear in neurons isolated from these mice. Since neuronal cells are particularly vulnerable to disturbances in microtubule dynamics, this mechanism might explain the neuron-specific CMT phenotype caused by HSPB1 mutations.


Assuntos
Proteínas de Choque Térmico HSP27/genética , Microtúbulos/metabolismo , Mutação/genética , Neurônios/metabolismo , Análise de Variância , Animais , Células Cultivadas , Chlorocebus aethiops , Gânglios Espinais/citologia , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Choque Térmico , Humanos , Gelo/efeitos adversos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/genética , Chaperonas Moleculares , Neurônios/efeitos dos fármacos , Nocodazol/farmacologia , Ligação Proteica , Ressonância de Plasmônio de Superfície , Espectrometria de Massas em Tandem/métodos , Fatores de Tempo , Transfecção/métodos , Tubulina (Proteína)/genética , Tubulina (Proteína)/farmacologia , Moduladores de Tubulina/farmacologia
14.
Hum Mol Genet ; 19(16): 3254-65, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20538880

RESUMO

Missense mutations (K141N and K141E) in the alpha-crystallin domain of the small heat shock protein HSPB8 (HSP22) cause distal hereditary motor neuropathy (distal HMN) or Charcot-Marie-Tooth neuropathy type 2L (CMT2L). The mechanism through which mutant HSPB8 leads to a specific motor neuron disease phenotype is currently unknown. To address this question, we compared the effect of mutant HSPB8 in primary neuronal and glial cell cultures. In motor neurons, expression of both HSPB8 K141N and K141E mutations clearly resulted in neurite degeneration, as manifested by a reduction in number of neurites per cell, as well as in a reduction in average length of the neurites. Furthermore, expression of the K141E (and to a lesser extent, K141N) mutation also induced spheroids in the neurites. We did not detect any signs of apoptosis in motor neurons, showing that mutant HSPB8 resulted in neurite degeneration without inducing neuronal death. While overt in motor neurons, these phenotypes were only very mildly present in sensory neurons and completely absent in cortical neurons. Also glial cells did not show an altered phenotype upon expression of mutant HSPB8. These findings show that despite the ubiquitous presence of HSPB8, only motor neurons appear to be affected by the K141N and K141E mutations which explain the predominant motor neuron phenotype in distal HMN and CMT2L.


Assuntos
Proteínas de Choque Térmico HSP20/metabolismo , Neurônios Motores/metabolismo , Proteínas Musculares/metabolismo , Mutação , Neuritos/metabolismo , Substituição de Aminoácidos , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Apoptose , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Dano ao DNA , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSP20/genética , Proteínas de Choque Térmico , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Chaperonas Moleculares , Neurônios Motores/patologia , Proteínas Musculares/genética , Neuritos/patologia , Neuroglia/metabolismo , Ratos , Ratos Wistar , Transfecção
15.
Sci Rep ; 10(1): 8755, 2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32472079

RESUMO

Three missense mutations targeting the same proline 209 (Pro209) codon in the co-chaperone Bcl2-associated athanogene 3 (BAG3) have been reported to cause distal myopathy, dilated cardiomyopathy or Charcot-Marie-Tooth type 2 neuropathy. Yet, it is unclear whether distinct molecular mechanisms underlie the variable clinical spectrum of the rare patients carrying these three heterozygous Pro209 mutations in BAG3. Here, we studied all three variants and compared them to the BAG3_Glu455Lys mutant, which causes dilated cardiomyopathy. We found that all BAG3_Pro209 mutants have acquired a toxic gain-of-function, which causes these variants to accumulate in the form of insoluble HDAC6- and vimentin-positive aggresomes. The aggresomes formed by mutant BAG3 led to a relocation of other chaperones such as HSPB8 and Hsp70, which, together with BAG3, promote the so-called chaperone-assisted selective autophagy (CASA). As a consequence of their increased aggregation-proneness, mutant BAG3 trapped ubiquitinylated client proteins at the aggresome, preventing their efficient clearance. Combined, these data show that all BAG3_Pro209 mutants, irrespective of their different clinical phenotypes, are characterized by a gain-of-function that contributes to the gradual loss of protein homeostasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Cardiomiopatia Dilatada/genética , Doença de Charcot-Marie-Tooth/genética , Miopatias Distais/genética , Chaperonas Moleculares/metabolismo , Mutação de Sentido Incorreto , Agregados Proteicos , Agregação Patológica de Proteínas/genética , Autofagia , Códon , Células HEK293 , Humanos , Prolina , Agregação Patológica de Proteínas/metabolismo , Transporte Proteico , Ubiquitinação
16.
Autophagy ; 15(6): 1051-1068, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30669930

RESUMO

HSPB1 (heat shock protein family B [small] member 1) is a ubiquitously expressed molecular chaperone. Most mutations in HSPB1 cause axonal Charcot-Marie-Tooth neuropathy and/or distal hereditary motor neuropathy. In this study we show that mutations in HSPB1 lead to impairment of macroautophagic/autophagic flux. In HSPB1 knockout cells, we demonstrate that HSPB1 is necessary for autophagosome formation, which was rescued upon re-expression of HSPB1. Employing a label-free LC-MS/MS analysis on the various HSPB1 variants (wild type and mutants), we identified autophagy-specific interactors. We reveal that the wild-type HSPB1 protein binds to the autophagy receptor SQSTM1/p62 and that the PB1 domain of SQSTM1 is essential for this interaction. Mutations in HSPB1 lead to a decrease in the formation of SQSTM1/p62 bodies, and subsequent impairment of phagophore formation, suggesting a regulatory role for HSPB1 in autophagy via interaction with SQSTM1. Remarkably, autophagy deficits could also be confirmed in patient-derived motor neurons thereby indicating that the impairment of autophagy might be one of the pathomechanisms by which mutations in HSPB1 lead to peripheral neuropathy. Abbreviations: ACD: alpha-crystallin domain; ALS: amyotrophic lateral sclerosis; ATG14: autophagy related 14; BAG1/3: BCL2 associated athanogene 1/3; CMT: Charcot-Marie-Tooth; dHMN: distal hereditary motor neuropathy; GFP: green fluorescent protein; HSPA8: heat shock protein family A (Hsp70) member 8; HSPB1/6/8: heat shock protein family B (small) member 1/6/8; LIR: LC3-interacting region; LC3B: microtubule associated protein 1 light chain 3 beta; PB1: Phox and Bem1; SQSTM1: sequestosome 1; STUB1/CHIP: STIP1 homology and U-box containing protein 1; UBA: ubiquitin-associated; WIPI1: WD repeat domain, phosphoinositide interacting 1; WT: wild-type.


Assuntos
Autofagossomos/metabolismo , Doença de Charcot-Marie-Tooth/genética , Proteínas de Choque Térmico/genética , Chaperonas Moleculares/genética , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Proteína Sequestossoma-1/metabolismo , Esclerose Lateral Amiotrófica/genética , Autofagossomos/ultraestrutura , Autofagia/genética , Cromatografia Líquida , Células HeLa , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Neurônios Motores/patologia , Mutação , Domínios Proteicos , Proteína Sequestossoma-1/química , Proteína Sequestossoma-1/genética , Espectrometria de Massas em Tandem
17.
Nat Commun ; 10(1): 5045, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31695036

RESUMO

Charcot-Marie-Tooth disease (CMT) is a length-dependent peripheral neuropathy. The aminoacyl-tRNA synthetases constitute the largest protein family implicated in CMT. Aminoacyl-tRNA synthetases are predominantly cytoplasmic, but are also present in the nucleus. Here we show that a nuclear function of tyrosyl-tRNA synthetase (TyrRS) is implicated in a Drosophila model of CMT. CMT-causing mutations in TyrRS induce unique conformational changes, which confer capacity for aberrant interactions with transcriptional regulators in the nucleus, leading to transcription factor E2F1 hyperactivation. Using neuronal tissues, we reveal a broad transcriptional regulation network associated with wild-type TyrRS expression, which is disturbed when a CMT-mutant is expressed. Pharmacological inhibition of TyrRS nuclear entry with embelin reduces, whereas genetic nuclear exclusion of mutant TyrRS prevents hallmark phenotypes of CMT in the Drosophila model. These data highlight that this translation factor may contribute to transcriptional regulation in neurons, and suggest a therapeutic strategy for CMT.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Núcleo Celular/metabolismo , Doença de Charcot-Marie-Tooth/metabolismo , Predisposição Genética para Doença , Aminoacil-tRNA Sintetases/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal , Núcleo Celular/enzimologia , Doença de Charcot-Marie-Tooth/genética , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/metabolismo , Feminino , Células HEK293 , Humanos , Larva , Masculino , Mutação , Doenças do Sistema Nervoso , Junção Neuromuscular , Neurônios/metabolismo , Fenótipo , Fatores de Transcrição/metabolismo
18.
Acta Neuropathol Commun ; 5(1): 5, 2017 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28077174

RESUMO

The small heat shock protein HSPB1 (Hsp27) is an ubiquitously expressed molecular chaperone able to regulate various cellular functions like actin dynamics, oxidative stress regulation and anti-apoptosis. So far disease causing mutations in HSPB1 have been associated with neurodegenerative diseases such as distal hereditary motor neuropathy, Charcot-Marie-Tooth disease and amyotrophic lateral sclerosis. Most mutations in HSPB1 target its highly conserved α-crystallin domain, while other mutations affect the C- or N-terminal regions or its promotor. Mutations inside the α-crystallin domain have been shown to enhance the chaperone activity of HSPB1 and increase the binding to client proteins. However, the HSPB1-P182L mutation, located outside and downstream of the α-crystallin domain, behaves differently. This specific HSPB1 mutation results in a severe neuropathy phenotype affecting exclusively the motor neurons of the peripheral nervous system. We identified that the HSPB1-P182L mutant protein has a specifically increased interaction with the RNA binding protein poly(C)binding protein 1 (PCBP1) and results in a reduction of its translational repressive activity. RNA immunoprecipitation followed by RNA sequencing on mouse brain lead to the identification of PCBP1 mRNA targets. These targets contain larger 3'- and 5'-UTRs than average and are enriched in an RNA motif consisting of the CTCCTCCTCCTCC consensus sequence. Interestingly, next to the clear presence of neuronal transcripts among the identified PCBP1 targets we identified known genes associated with hereditary peripheral neuropathies and hereditary spastic paraplegias. We therefore conclude that HSPB1 can mediate translational repression through interaction with an RNA binding protein further supporting its role in neurodegenerative disease.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/genética , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Sequência Consenso , Proteínas de Ligação a DNA , Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Proteínas de Choque Térmico HSP27/genética , Células HeLa , Proteínas de Choque Térmico/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Camundongos , Chaperonas Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Proteínas de Neoplasias/genética , Ligação Proteica , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Regiões não Traduzidas
19.
Neurology ; 88(6): 533-542, 2017 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-28077491

RESUMO

OBJECTIVE: To identify the unknown genetic cause in a nuclear family with an axonal form of peripheral neuropathy and atypical disease course. METHODS: Detailed neurologic, electrophysiologic, and neuropathologic examinations of the patients were performed. Whole exome sequencing of both affected individuals was done. The effect of the identified sequence variations was investigated at cDNA and protein level in patient-derived lymphoblasts. The plasma sphingoid base profile was analyzed. Functional consequences of neuron-specific downregulation of the gene were studied in Drosophila. RESULTS: Both patients present an atypical form of axonal peripheral neuropathy, characterized by acute or subacute onset and episodes of recurrent mononeuropathy. We identified compound heterozygous mutations cosegregating with disease and absent in controls in the SGPL1 gene, encoding sphingosine 1-phosphate lyase (SPL). The p.Ser361* mutation triggers nonsense-mediated mRNA decay. The missense p.Ile184Thr mutation causes partial protein degradation. The plasma levels of sphingosine 1-phosphate and sphingosine/sphinganine ratio were increased in the patients. Neuron-specific downregulation of the Drosophila orthologue impaired the morphology of the neuromuscular junction and caused progressive degeneration of the chemosensory neurons innervating the wing margin bristles. CONCLUSIONS: We suggest SPL deficiency as a cause of a distinct form of Charcot-Marie-Tooth disease in humans, thus extending the currently recognized clinical and genetic spectrum of inherited peripheral neuropathies. Our data emphasize the importance of sphingolipid metabolism for neuronal function.


Assuntos
Aldeído Liases/deficiência , Aldeído Liases/genética , Aldeído Liases/metabolismo , Doença de Charcot-Marie-Tooth/genética , Códon sem Sentido , Proteínas de Drosophila/metabolismo , Mutação de Sentido Incorreto , Adulto , Animais , Animais Geneticamente Modificados , Células Cultivadas , Doença de Charcot-Marie-Tooth/fisiopatologia , Estudos de Coortes , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Lisofosfolipídeos/sangue , Masculino , Neurônios/metabolismo , Neurônios/patologia , Irmãos , Esfingosina/análogos & derivados , Esfingosina/sangue
20.
Neuromuscul Disord ; 22(8): 699-711, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22595202

RESUMO

Missense mutations in the small heat shock protein HSPB8 cause distal hereditary motor neuropathy (dHMN) and axonal Charcot-Marie-Tooth disease (CMT2L). We previously demonstrated that, despite the ubiquitous expression of HSPB8, motor neurons appear to be predominantly affected by HSPB8 mutations. Here, we studied the effect of mutant HSPB8 in primary fibroblast cultures derived from dHMN patients' skin biopsy. In early passage cultures, we observed in all patients' fibroblasts HSPB8 protein aggregates that were not detected in control cells. After applying heat shock stress on the patients' early passage cultured cells, the protein aggregates coalesced into larger formations, while in control cells a homogenous upregulation of HSPB8 protein expression was seen. We also found a reduction in the mitochondrial membrane potential in the early passage cultures. After three months in culture, the number of cells with aggregates had become indistinguishable from that in controls and the mitochondrial membrane potential had returned to normal. These results emphasize the possible drawbacks of using patients' non-neuronal cells to study neuropathological disease mechanisms.


Assuntos
Fibroblastos/fisiologia , Proteínas de Choque Térmico/genética , Potencial da Membrana Mitocondrial/fisiologia , Doença dos Neurônios Motores/patologia , Doença dos Neurônios Motores/fisiopatologia , Mutação de Sentido Incorreto/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas/metabolismo , Adulto , Idoso , Apoptose , Axônios/patologia , Biomarcadores/metabolismo , Biópsia , Células Cultivadas , Feminino , Fibroblastos/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares , Ligação Proteica , Pele/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA