Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 19(1)2018 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-29316663

RESUMO

The maintenance of proteome integrity is of primary importance in post-mitotic tissues such as muscle cells; thus, protein quality control mechanisms must be carefully regulated to ensure their optimal efficiency, a failure of these processes being associated with various muscular disorders. Duchenne muscular dystrophy (DMD) is one of the most common and severe forms of muscular dystrophies and is caused by mutations in the dystrophin gene. Protein quality control modulations have been diversely observed in degenerating muscles of patients suffering from DMD or in animal models of the disease. In this study, we investigated whether modulations of protein quality control mechanisms already pre-exist in undifferentiated myoblasts originating from DMD patients. We report for the first time that the absence of dystrophin in human myoblasts is associated with protein aggregation stress characterized by an increase of protein aggregates. This stress is combined with BAG1 to BAG3 switch, NFκB activation and up-regulation of BAG3/HSPB8 complexes that ensure preferential routing of misfolded/aggregated proteins to autophagy rather than to deficient 26S proteasome. In this context, restoration of pre-existing alterations of protein quality control processes might represent an alternative strategy for DMD therapies.


Assuntos
Autofagia , Distrofia Muscular de Duchenne/metabolismo , Mioblastos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Resposta a Proteínas não Dobradas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Humanos , Mioblastos/patologia , NF-kappa B/metabolismo , Fatores de Transcrição/metabolismo
2.
Hum Mol Genet ; 24(22): 6428-45, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26358775

RESUMO

Duchenne muscular dystrophy (DMD) is a genetic disease characterized by progressive muscle degeneration due to mutations in the dystrophin gene. In spite of great advances in the design of curative treatments, most patients currently receive palliative therapies with steroid molecules such as prednisone or deflazacort thought to act through their immunosuppressive properties. These molecules only slightly slow down the progression of the disease and lead to severe side effects. Fundamental research is still needed to reveal the mechanisms involved in the disease that could be exploited as therapeutic targets. By studying a Caenorhabditis elegans model for DMD, we show here that dystrophin-dependent muscle degeneration is likely to be cell autonomous and affects the muscle cells the most involved in locomotion. We demonstrate that muscle degeneration is dependent on exercise and force production. Exhaustive studies by electron microscopy allowed establishing for the first time the chronology of subcellular events occurring during the entire process of muscle degeneration. This chronology highlighted the crucial role for dystrophin in stabilizing sarcomeric anchoring structures and the sarcolemma. Our results suggest that the disruption of sarcomeric anchoring structures and sarcolemma integrity, observed at the onset of the muscle degeneration process, triggers subcellular consequences that lead to muscle cell death. An ultra-structural analysis of muscle biopsies from DMD patients suggested that the chronology of subcellular events established in C. elegans models the pathogenesis in human. Finally, we found that the loss of sarcolemma integrity was greatly reduced after prednisone treatment suggesting a role for this molecule in plasma membrane stabilization.


Assuntos
Distrofia Muscular de Duchenne/patologia , Sarcolema/ultraestrutura , Sarcômeros/patologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animais de Doenças , Distrofina/genética , Distrofina/metabolismo , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Mutação , Sarcolema/metabolismo , Sarcolema/patologia , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura
3.
Hum Mol Genet ; 22(22): 4562-78, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23804750

RESUMO

Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by mutations in the dystrophin gene. The subcellular mechanisms of DMD remain poorly understood and there is currently no curative treatment available. Using a Caenorhabditis elegans model for DMD as a pharmacologic and genetic tool, we found that cyclosporine A (CsA) reduces muscle degeneration at low dose and acts, at least in part, through a mitochondrial cyclophilin D, CYN-1. We thus hypothesized that CsA acts on mitochondrial permeability modulation through cyclophilin D inhibition. Mitochondrial patterns and dynamics were analyzed, which revealed dramatic mitochondrial fragmentation not only in dystrophic nematodes, but also in a zebrafish model for DMD. This abnormal mitochondrial fragmentation occurs before any obvious sign of degeneration can be detected. Moreover, we demonstrate that blocking/delaying mitochondrial fragmentation by knocking down the fission-promoting gene drp-1 reduces muscle degeneration and improves locomotion abilities of dystrophic nematodes. Further experiments revealed that cytochrome c is involved in muscle degeneration in C. elegans and seems to act, at least in part, through an interaction with the inositol trisphosphate receptor calcium channel, ITR-1. Altogether, our findings reveal that mitochondria play a key role in the early process of muscle degeneration and may be a target of choice for the design of novel therapeutics for DMD. In addition, our results provide the first indication in the nematode that (i) mitochondrial permeability transition can occur and (ii) cytochrome c can act in cell death.


Assuntos
Ciclofilinas/metabolismo , Ciclosporina/farmacologia , Citocromos c/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Dinâmica Mitocondrial , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Morte Celular , Ciclofilinas/antagonistas & inibidores , Citocromos c/genética , Técnicas de Silenciamento de Genes , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Metazolamida/farmacologia , Dinâmica Mitocondrial/efeitos dos fármacos , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Filogenia , Homologia de Sequência , Peixe-Zebra/embriologia , Peixe-Zebra/genética
4.
Methods ; 56(1): 103-13, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22041718

RESUMO

We previously reported the use of the cheap and fast-growing nematode Caenorhabditis elegans to search for molecules, which reduce muscle degeneration in a model for Duchenne Muscular Dystrophy (DMD). We showed that Prednisone, a steroid that is generally prescribed as a palliative treatment to DMD patients, also reduced muscle degeneration in the C. elegans DMD model. We further showed that this strategy could lead to the discovery of new and unsuspected small molecules, which have been further validated in a mammalian model of DMD, i.e. the mdx mouse model. These proof-of-principles demonstrate that C. elegans can serve as a screening tool to search for drugs against neuromuscular disorders. Here, we report and discuss two methodologies used to screen chemical libraries for drugs against muscle disorders in C. elegans. We first describe a manual method used to find drugs against DMD. We further present a semi-automated method, which is currently in use for the search of drugs against the Schwartz-Jampel Syndrome (SJS). Both assays are simple to implement and can be readily transposed and/or adapted to screens against other muscle/neuromuscular diseases, which can be modeled in the worm. Finally we discuss, with respect to our experience and knowledge, the different parameters that have to be taken into account before choosing one or the other method.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Automação , Modelos Animais de Doenças , Humanos , Distrofia Muscular Animal , Osteocondrodisplasias
5.
Sci Rep ; 10(1): 11665, 2020 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-32647220

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Aging Cell ; 17(2)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29314608

RESUMO

Aging is commonly defined as the loss of global homeostasis, which results from progressive alteration of all organs function. This model is currently challenged by recent data showing that interventions that extend lifespan do not always increase the overall fitness of the organism. These data suggest the existence of tissue-specific factors that regulate the pace of aging in a cell-autonomous manner. Here, we investigated aging of Caenorhabditis elegans striated muscles at the subcellular and the physiological level. Our data show that muscle aging is characterized by a dramatic decrease in the expression of genes encoding proteins required for muscle contraction, followed by a change in mitochondria morphology, and an increase in autophagosome number. Myofilaments, however, remain unaffected during aging. We demonstrated that the conserved transcription factor UNC-120/SRF regulates muscle aging biomarkers. Interestingly, the role of UNC-120/SRF in the control of muscle aging can be dissociated from its broader effect on lifespan. In daf-2/insulin/IGF1 receptor mutants, which exhibit a delayed appearance of muscle aging biomarkers and are long-lived, disruption of unc-120 accelerates muscle aging but does not suppress the lifespan phenotype of daf-2 mutant. Conversely, unc-120 overexpression delays muscle aging but does not increase lifespan. Overall, we demonstrate that UNC-120/SRF controls the pace of muscle aging in a cell-autonomous manner downstream of the insulin/IGF1 receptor.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Longevidade/genética , Proteínas de Domínio MADS/genética , Músculos/metabolismo , Envelhecimento , Animais , Proteínas de Caenorhabditis elegans/genética , Fatores de Transcrição
7.
Sci Rep ; 8(1): 7354, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743663

RESUMO

Mitochondria are double-membrane subcellular organelles with highly conserved metabolic functions including ATP production. Mitochondria shapes change continually through the combined actions of fission and fusion events rendering mitochondrial network very dynamic. Mitochondria are largely implicated in pathologies and mitochondrial dynamics is often disrupted upon muscle degeneration in various models. Currently, the exact roles of mitochondria in the molecular mechanisms that lead to muscle degeneration remain poorly understood. Here we report a role for DRP-1 in regulating apoptosis induced by dystrophin-dependent muscle degeneration. We found that: (i) dystrophin-dependent muscle degeneration was accompanied by a drastic increase in mitochondrial fragmentation that can be rescued by genetic manipulations of mitochondrial dynamics (ii) the loss of function of the fission gene drp-1 or the overexpression of the fusion genes eat-3 and fzo-1 provoked a reduction of muscle degeneration and an improved mobility of dystrophin mutant worms (iii) the functions of DRP-1 in apoptosis and of others apoptosis executors are important for dystrophin-dependent muscle cell death (iv) DRP-1-mediated apoptosis is also likely to induce age-dependent loss of muscle cell. Collectively, our findings point toward a mechanism involving mitochondrial dynamics to respond to trigger(s) of muscle degeneration via apoptosis in Caenorhabditis elegans.


Assuntos
Apoptose/genética , Proteínas de Caenorhabditis elegans/metabolismo , Dinaminas/metabolismo , Distrofina/genética , Músculos/metabolismo , Mutação , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caspases/metabolismo , Locomoção/genética , Mitocôndrias/metabolismo , Dinâmica Mitocondrial
8.
J Mol Biol ; 358(2): 387-95, 2006 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-16527307

RESUMO

The Caenorhabditis elegans SLO-1 channel belongs to the family of calcium-activated large conductance BK potassium channels. SLO-1 has been shown to be involved in neurotransmitter release and ethanol response. Here, we report that SLO-1 also has a critical role in muscles. Inactivation of the slo-1 gene in muscles leads to phenotypes similar to those caused by mutations of the dystrophin homologue dys-1. Notably, slo-1 mutations result in a progressive muscle degeneration when put into a sensitized genetic background. slo-1 localization was observed by gfp reporter gene in both the M-line and the dense bodies (Z line) of the C.elegans body-wall muscles. Using the inside-out configuration of the patch clamp technique on body-wall muscle cells of acutely dissected wild-type worms, we characterized a Ca2+-activated K+ channel that was identified unambiguously as SLO-1. Since neither the abundance nor the conductance of SLO-1 was changed significantly in dys-1 mutants compared to wild-type animals, it is likely that the inactivation of dys-1 causes a misregulation of SLO-1. All in all, these results indicate that SLO-1 function in C.elegans muscles is related to the dystrophin homologue DYS-1.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Distrofina/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Músculos/metabolismo , Distrofia Muscular Animal/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans/metabolismo , Músculos/patologia , Distrofia Muscular Animal/patologia , Mutação/genética , Fenótipo , Canais de Potássio Cálcio-Ativados/metabolismo
9.
WormBook ; 2017: 1-59, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-27555356

RESUMO

In C. elegans, mutants that are defective in muscle function and/or structure are easy to detect and analyze since: 1) body wall muscle is essential for locomotion, and 2) muscle structure can be assessed by multiple methods including polarized light, electron microscopy (EM), Green Fluorescent Protein (GFP) tagged proteins, and immunofluorescence microscopy. The overall structure of the sarcomere, the fundamental unit of contraction, is conserved from C. elegans to man, and the molecules involved in sarcomere assembly, maintenance, and regulation of muscle contraction are also largely conserved. This review reports the latest findings on the following topics: the transcriptional network that regulates muscle differentiation, identification/function/dynamics of muscle attachment site proteins, regulation of the assembly and maintenance of the sarcomere by chaperones and proteases, the role of muscle-specific giant protein kinases in sarcomere assembly, and the regulation of contractile activity, and new insights into the functions of the dystrophin glycoprotein complex.


Assuntos
Caenorhabditis elegans/fisiologia , Desenvolvimento Muscular , Músculos , Animais , Caenorhabditis elegans/anatomia & histologia , Caenorhabditis elegans/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Chaperonas Moleculares , Desenvolvimento Muscular/genética , Proteínas Musculares/fisiologia , Músculos/anatomia & histologia , Músculos/embriologia , Músculos/fisiologia , Sarcômeros/fisiologia
10.
J Mol Biol ; 332(5): 1037-46, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-14499607

RESUMO

Syntrophins are a family of PDZ domain-containing adaptor proteins required for receptor localization. Syntrophins are also associated with the dystrophin complex in muscles. We report here the molecular and functional characterization of the Caenorhabditis elegans gene stn-1 (F30A10.8), which encodes a syntrophin with homology to vertebrate alpha and beta-syntrophins. stn-1 is expressed in neurons and in muscles of C.elegans. stn-1 mutants resemble dystrophin (dys-1) and dystrobrevin (dyb-1) mutants: they are hyperactive, bend their heads when they move forward, tend to hypercontract, and are hypersensitive to the acetylcholinesterase inhibitor aldicarb. These phenotypes are suppressed when stn-1 is expressed under the control of a muscular promoter, indicating that they are caused by the absence of stn-1 in muscles. These results suggest that the role of syntrophin is linked to dystrophin function in C.elegans.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas Associadas à Distrofina , Distrofina/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neuropeptídeos/fisiologia , Aldicarb/farmacologia , Animais , Caenorhabditis elegans , Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio , Bases de Dados como Assunto , Genoma , Genótipo , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Modelos Biológicos , Músculos/patologia , Distrofia Muscular de Duchenne/metabolismo , Mutação , Fenótipo , Filogenia , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Sensibilidade e Especificidade , Técnicas do Sistema de Duplo-Híbrido
11.
Gene ; 294(1-2): 77-86, 2002 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-12234669

RESUMO

Dystrophin is the product of the gene mutated in Duchenne muscular dystrophy (DMD). Neither the function of dystrophin nor the physiopathology of the disease have been clearly established so far. In mammals, the dystrophin-glycoprotein complex (DGC) includes dystrophin, as well as transmembrane and cytoplasmic proteins. Since Caenorhabditis elegans possesses a dystrophin-like gene (dys-1), we investigated whether homologues of the DGC members could also be found in the C. elegans genome. Conserved homologues were found for dystroglycan, delta/gamma-sarcoglycan and syntrophin. Divergent but related proteins were found for alpha- and beta-sarcoglycans. No sarcospan counterpart was found. The expression of the conserved homologues was inactivated using the RNA interference technique. Phenotypes similar to that of dys-1 were obtained, both in the wild-type background and in combination with other mutations. These results strongly suggest that a protein complex comprising functional analogies with the DGC exists in C. elegans.


Assuntos
Caenorhabditis elegans/genética , Proteínas Associadas à Distrofina , Distrofina/genética , Glicoproteínas de Membrana/genética , Sequência de Aminoácidos , Animais , Proteínas do Citoesqueleto/genética , Distroglicanas , Proteínas de Membrana/genética , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Musculares/genética , Músculos/anormalidades , Músculos/efeitos dos fármacos , Músculos/metabolismo , Interferência de RNA , RNA de Cadeia Dupla/administração & dosagem , RNA de Cadeia Dupla/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
12.
Neuromuscul Disord ; 12(4): 371-7, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12062255

RESUMO

Duchenne muscular dystrophy is one of the most common neuromuscular diseases. It is caused by mutations in the dystrophin gene. Dystrobrevins are dystrophin-associated proteins potentially involved in signal transduction. The nematode Caenorhabditis elegans possesses one dystrophin-like (dys-1) and one dystrobrevin-like (dyb-1) gene. Mutations of dyb-1 and dys-1 lead to similar phenotypes, comprising hyperactivity and a tendency to hypercontract, which suggest that these proteins may participate in a common function. We show here that overexpression of the Dyb-1 protein delays the onset of the myopathy observed in the C. elegans double mutant (dys-1; hlh-1 mutations). This finding indicates that, in C. elegans, (1) the absence of dystrophin can be partly compensated for by extra doses of dystrobrevin, and (2) dystrobrevin is partly functional in absence of dystrophin.


Assuntos
Proteínas de Caenorhabditis elegans , Distrofina/genética , Locomoção/genética , Proteínas do Tecido Nervoso , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Animais , Caenorhabditis elegans , Modelos Animais de Doenças , Expressão Gênica/fisiologia , Músculos/metabolismo , Músculos/patologia , Distrofia Muscular de Duchenne/genética , Fenótipo
13.
J Vis Exp ; (82): e50773, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24379027

RESUMO

Microinjecting DNA into the cytoplasm of the syncytial gonad of Caenorhabditis elegans is the main technique used to establish transgenic lines that exhibit partial and variable transmission rates of extrachromosomal arrays to the next generation. In addition, transgenic animals are mosaic and express the transgene in a variable number of cells. Extrachromosomal arrays can be integrated into the C. elegans genome using UV irradiation to establish nonmosaic transgenic strains with 100% transmission rate of the transgene. To that extent, F1 progenies of UV irradiated transgenic animals are screened for animals carrying a heterozygous integration of the transgene, which leads to a 75% Mendelian transmission rate to the F2 progeny. One of the challenges of this method is to distinguish between the percentage of transgene transmission in a population before (X% transgenic animals) and after integration (≥75% transgenic F2 animals). Thus, this method requires choosing a nonintegrated transgenic line with a percentage of transgenic animals that is significantly lower than the Mendelian segregation of 75%. Consequently, nonintegrated transgenic lines with an extrachromosomal array transmission rate to the next generation ≤60% are usually preferred for integration, and transgene integration in highly transmitting strains is difficult. Here we show that the efficiency of extrachromosomal arrays integration into the genome is increased when using highly transmitting transgenic lines (≥80%). The described protocol allows for easy selection of several independent lines with homozygous transgene integration into the genome after UV irradiation of transgenic worms exhibiting a high rate of extrachromosomal array transmission. Furthermore, this method is quite fast and low material consuming. The possibility of rapidly generating different lines that express a particular integrated transgene is of great interest for studies focusing on gene expression pattern and regulation, protein localization, and overexpression, as well as for the development of subcellular markers.


Assuntos
Caenorhabditis elegans/genética , Cromossomos , Genoma Helmíntico , Transgenes , Animais , Animais Geneticamente Modificados , DNA/administração & dosagem , DNA/genética , Microinjeções
14.
Mol Biol Cell ; 24(8): 1232-49, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23427270

RESUMO

In vertebrates, zyxin is a LIM-domain protein belonging to a family composed of seven members. We show that the nematode Caenorhabditis elegans has a unique zyxin-like protein, ZYX-1, which is the orthologue of the vertebrate zyxin subfamily composed of zyxin, migfilin, TRIP6, and LPP. The ZYX-1 protein is expressed in the striated body-wall muscles and localizes at dense bodies/Z-discs and M-lines, as well as in the nucleus. In yeast two-hybrid assays ZYX-1 interacts with several known dense body and M-line proteins, including DEB-1 (vinculin) and ATN-1 (α-actinin). ZYX-1 is mainly localized in the middle region of the dense body/Z-disk, overlapping the apical and basal regions containing, respectively, ATN-1 and DEB-1. The localization and dynamics of ZYX-1 at dense bodies depend on the presence of ATN-1. Fluorescence recovery after photobleaching experiments revealed a high mobility of the ZYX-1 protein within muscle cells, in particular at dense bodies and M-lines, indicating a peripheral and dynamic association of ZYX-1 at these muscle adhesion structures. A portion of the ZYX-1 protein shuttles from the cytoplasm into the nucleus, suggesting a role for ZYX-1 in signal transduction. We provide evidence that the zyx-1 gene encodes two different isoforms, ZYX-1a and ZYX-1b, which exhibit different roles in dystrophin-dependent muscle degeneration occurring in a C. elegans model of Duchenne muscular dystrophy.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Distrofina/metabolismo , Músculos/metabolismo , Zixina/fisiologia , Actinina/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/citologia , Proteínas de Caenorhabditis elegans/química , Expressão Gênica , Dados de Sequência Molecular , Músculos/citologia , Especificidade de Órgãos , Filogenia , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Transporte Proteico , Homologia de Sequência de Aminoácidos , Zixina/química
15.
PLoS One ; 7(2): e30482, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22347378

RESUMO

Methods that use homologous recombination to engineer the genome of C. elegans commonly use strains carrying specific insertions of the heterologous transposon Mos1. A large collection of known Mos1 insertion alleles would therefore be of general interest to the C. elegans research community. We describe here the optimization of a semi-automated methodology for the construction of a substantial collection of Mos1 insertion mutant strains. At peak production, more than 5,000 strains were generated per month. These strains were then subject to molecular analysis, and more than 13,300 Mos1 insertions characterized. In addition to targeting directly more than 4,700 genes, these alleles represent the potential starting point for the engineered deletion of essentially all C. elegans genes and the modification of more than 40% of them. This collection of mutants, generated under the auspices of the European NEMAGENETAG consortium, is publicly available and represents an important research resource.


Assuntos
Caenorhabditis elegans/genética , Elementos de DNA Transponíveis , Proteínas de Ligação a DNA , Engenharia Genética/métodos , Genoma/genética , Recombinação Genética , Transposases , Animais , Animais Geneticamente Modificados , Recombinação Homóloga , Mutagênese Insercional , Pesquisa
16.
Mol Biol Cell ; 19(3): 785-96, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094057

RESUMO

In Caenorhabditis elegans, mutations of the dystrophin homologue, dys-1, produce a peculiar behavioral phenotype (hyperactivity and a tendency to hypercontract). In a sensitized genetic background, dys-1 mutations also lead to muscle necrosis. The dyc-1 gene was previously identified in a genetic screen because its mutation leads to the same phenotype as dys-1, suggesting that the two genes are functionally linked. Here, we report the detailed characterization of the dyc-1 gene. dyc-1 encodes two isoforms, which are expressed in neurons and muscles. Isoform-specific RNAi experiments show that the absence of the muscle isoform, and not that of the neuronal isoform, is responsible for the dyc-1 mutant phenotype. In the sarcomere, the DYC-1 protein is localized at the edges of the dense body, the nematode muscle adhesion structure where actin filaments are anchored and linked to the sarcolemma. In yeast two-hybrid assays, DYC-1 interacts with ZYX-1, the homologue of the vertebrate focal adhesion LIM domain protein zyxin. ZYX-1 localizes at dense bodies and M-lines as well as in the nucleus of C. elegans striated muscles. The DYC-1 protein possesses a highly conserved 19 amino acid sequence, which is involved in the interaction with ZYX-1 and which is sufficient for addressing DYC-1 to the dense body. Altogether our findings indicate that DYC-1 may be involved in dense body function and stability. This, taken together with the functional link between the C. elegans DYC-1 and DYS-1 proteins, furthermore suggests a requirement of dystrophin function at this structure. As the dense body shares functional similarity with both the vertebrate Z-disk and the costamere, we therefore postulate that disruption of muscle cell adhesion structures might be the primary event of muscle degeneration occurring in the absence of dystrophin, in C. elegans as well as vertebrates.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Proteínas de Transporte/metabolismo , Estruturas Citoplasmáticas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Distrofina/metabolismo , Proteínas Musculares/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/ultraestrutura , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Sequência Conservada , Estruturas Citoplasmáticas/ultraestrutura , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Regulação da Expressão Gênica , Proteínas com Domínio LIM , Microscopia Imunoeletrônica , Modelos Biológicos , Dados de Sequência Molecular , Músculos/citologia , Músculos/metabolismo , Mutação/genética , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares/metabolismo , Zixina
17.
J Muscle Res Cell Motil ; 28(1): 79-87, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17492481

RESUMO

During evolution, both the architecture and the cellular physiology of muscles have been remarkably maintained. Striated muscles of invertebrates, although less complex, strongly resemble vertebrate skeletal muscles. In particular, the basic contractile unit called the sarcomere is almost identical between vertebrates and invertebrates. In vertebrate muscles, sarcomeric actin filaments are anchored to attachment points called Z-disks, which are linked to the extra-cellular matrix (ECM) by a muscle specific focal adhesion site called the costamere. In this review, we focus on the dense body of the animal model Caenorhabditis elegans. The C. elegans dense body is a structure that performs two in one roles at the same time, that of the Z-disk and of the costamere. The dense body is anchored in the muscle membrane and provides rigidity to the muscle by mechanically linking actin filaments to the ECM. In the last few years, it has become increasingly evident that, in addition to its structural role, the dense body also performs a signaling function in muscle cells. In this paper, we review recent advances in the understanding of the C. elegans dense body composition and function.


Assuntos
Caenorhabditis elegans/ultraestrutura , Estruturas Citoplasmáticas/metabolismo , Proteínas Musculares/metabolismo , Músculos/metabolismo , Músculos/ultraestrutura , Citoesqueleto de Actina/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Estruturas Citoplasmáticas/ultraestrutura , Sarcolema/ultraestrutura , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Transdução de Sinais
18.
Eur J Biochem ; 269(6): 1607-12, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11895430

RESUMO

Dystrobrevin is one of the intracellular components of the transmembrane dystrophin-glycoprotein complex (DGC). The functional role of this complex in normal and pathological situations has not yet been clearly established. Dystrobrevin disappears from the muscle membrane in Duchenne muscular dystrophy (DMD), which results from dystrophin mutations, as well as in limb girdle muscular dystrophies (LGMD), which results from mutations affecting other members of the DGC complex. These findings therefore suggest that dystrobrevin may play a pivotal role in the progression of these clinically related diseases. In this study, we used the Caenorhabditis elegans model to address the question of the relationship between dystrobrevin binding to dystrophin and dystrobrevin function. Deletions of the dystrobrevin protein were performed and the ability of the mutated forms to bind to dystrophin was tested both in vitro and in a two-hybrid assay, as well as their ability to rescue dystrobrevin (dyb-1) mutations in C. elegans. The deletions affecting the second helix of the Dyb-1 coiled-coil domain abolished the binding of dystrobrevin to dystrophin both in vitro and in the two-hybrid assay. These deletions also abolished the rescuing activity of a functional transgene in vivo. These results are consistent with a model according to which dystrobrevin must bind to dystrophin to be able to function properly.


Assuntos
Caenorhabditis elegans/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Associadas à Distrofina , Distrofina/metabolismo , Proteínas de Membrana/metabolismo , Animais , Sítios de Ligação , Caenorhabditis elegans/genética , Proteínas do Citoesqueleto/química , Teste de Complementação Genética , Proteínas de Membrana/química , Mutagênese , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA