Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Exp Neurol ; 232(2): 185-94, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21906591

RESUMO

Store-operated Ca(2+) entry (SOCE) over the plasma membrane is activated by depletion of intracellular Ca(2+) stores and has only recently been shown to play a role in CNS processes like synaptic plasticity. However, the direct effect of SOCE on the excitability of neuronal networks in vitro and in vivo has never been determined. We confirmed the presence of SOCE and the expression of the calcium sensors STIM1 and STIM2, which convey information about the calcium load of the stores to channel proteins at the plasma membrane, in neurons and astrocytes. Inhibition of SOCE by pharmacological agents 2-APB and ML-9 reduced the steady-state neuronal Ca(2+) concentration, reduced network activity, and increased synchrony of primary neuronal cultures grown on multi-electrode arrays, which prompted us to elucidate the relative expression of STIM proteins in conditions of pathologic excitability. Both proteins were increased in brains of chronic epileptic rodents and strongly expressed in hippocampal specimens from medial temporal lobe epilepsy patients. Pharmacologic inhibition of SOCE in chronic epileptic hippocampal slices suppressed interictal spikes and rhythmized epileptic burst activity. Our results indicate that SOCE modulates the activity of neuronal networks in vitro and in vivo and delineates SOCE as a potential drug target.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Moléculas de Adesão Celular/metabolismo , Membrana Celular/metabolismo , Doença Crônica , Córtex Entorrinal/citologia , Córtex Entorrinal/fisiopatologia , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/citologia , Hipocampo/fisiopatologia , Humanos , Proteínas de Neoplasias/metabolismo , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Neurônios/citologia , Técnicas de Cultura de Órgãos , Cultura Primária de Células , Ratos , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal
2.
Biochim Biophys Acta ; 1793(6): 1050-7, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19111578

RESUMO

Mutations in presenilin 1 (PS1), which are the major cause of familial Alzheimer's disease (FAD), are involved in perturbations of cellular Ca2+ homeostasis. Attenuation of capacitative Ca2+ entry (CCE) is the most often observed alteration of Ca2+ homeostasis in cells bearing FAD PS1 mutations. However, molecular mechanisms underlying this CCE impairment remains elusive. We demonstrate that cellular levels of STIM1 and STIM2 proteins, which are key players in CCE, depend on presenilins. We found increased level of STIM1 and decreased level of STIM2 proteins in mouse embryonic fibroblasts lacking presenilins. Fura-2 ratiometric assays revealed that CCE is enhanced in these cells after Ca2+ stores depletion by thapsigargin treatment. In turn, overexpression of PS1 with FAD mutations in HEK293 cells led to an attenuation of CCE. Although, no changes in STIM protein levels were observed in these HEK293 cells, FAD mutations in endogenous PS1 in human B lymphocytes resulted in a decreased expression of STIM2 in parallel to an attenuation of CCE. Our experiments showing that knock-out of presenilins in MEF cells and FAD mutations in endogenous PS1 in lymphocytes affect both CCE and the cellular level of STIM proteins open new perspectives for studies on CCE in FAD.


Assuntos
Doença de Alzheimer/metabolismo , Moléculas de Adesão Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Idoso , Doença de Alzheimer/genética , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Canais de Cálcio , Moléculas de Adesão Celular/genética , Células Cultivadas , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/genética , Presenilina-1/genética , Presenilina-2/genética , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal
3.
J Biol Chem ; 280(25): 23475-83, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15849365

RESUMO

The extracellular domain of the mature form of ADAM12 consists of the metalloprotease, disintegrin, cysteine-rich, and epidermal growth factor (EGF)-like domains. The disintegrin, cysteine-rich, and EGF-like fragments have been shown previously to support cell adhesion via activated integrins or proteoglycans. In this study, we report that the entire extracellular domain of mouse ADAM12 produced in Drosophila S2 cells supported efficient adhesion and spreading of C2C12 myoblasts even in the absence of exogenous integrin activators. This adhesion was not mediated by beta1 integrins or proteoglycans, was myoblast-specific, and required the presence of both the metalloprotease and disintegrin/cysteine-rich domains of ADAM12. Analysis of the recombinant proteins by far-UV circular dichroism suggested that the secondary structures of the autonomously expressed metalloprotease domain and the disintegrin/cysteine-rich/EGF-like domains differ from the structures present in the intact extracellular domain. Furthermore, the intact extracellular domain (but not the metalloprotease domain or the disintegrin/cysteine-rich/EGF-like fragment alone) decreased the expression of the cell cycle inhibitor p21 and myogenin, two markers of differentiation, and inhibited C2C12 myoblast fusion. Thus, the novel protein-protein interaction reported here involving the extracellular domain of ADAM12 may have important biological consequences during myoblast differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Musculares/fisiologia , Mioblastos/citologia , Células 3T3 , Proteínas ADAM , Proteína ADAM12 , Animais , Células CHO , Dicroísmo Circular , Cricetinae , Drosophila , Proteínas de Membrana/química , Camundongos , Proteínas Musculares/química
4.
Biochem J ; 385(Pt 1): 309-17, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15361073

RESUMO

The extracellular domain of integrin alpha7 is ADP-ribosylated by an arginine-specific ecto-ADP-ribosyltransferase after adding exogenous NAD+ to intact C2C12 skeletal muscle cells. The effect of ADP-ribosylation on the structure or function of integrin alpha7beta1 has not been explored. In the present study, we show that ADP-ribosylation of integrin alpha7 takes place exclusively in differentiated myotubes and that this post-translational modification modulates the affinity of alpha7beta1 dimer for its ligand, laminin. ADP-ribosylation in the 37-kDa 'stalk' region of alpha7 that takes place at micromolar NAD+ concentrations increases the binding of the alpha7beta1 dimer to laminin. Increased in vitro binding of integrin alpha7beta1 to laminin after ADP-ribosylation of the 37-kDa fragment of alpha7 requires the presence of Mn2+ and it is not observed in the presence of Mg2+. In contrast, ADP-ribosylation of the 63-kDa N-terminal region comprising the ligand-binding site of alpha7 that occurs at approx. 100 microM NAD+ inhibits the binding of integrin alpha7beta1 to laminin. Furthermore, incubation of C2C12 myotubes with NAD+ increases the expression of an epitope on integrin beta1 subunit recognized by monoclonal antibody 9EG7. We discuss our results based on the current models of integrin activation. We also hypothesize that ADP-ribosylation may represent a mechanism of regulation of integrin alpha7beta1 function in myofibres in vivo when the continuity of the membrane is compromised and NAD+ is available as a substrate for ecto-ADP-ribosylation.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Antígenos CD/metabolismo , Cadeias alfa de Integrinas/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Animais , Antígenos CD/química , Cátions Bivalentes/farmacologia , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Cadeias alfa de Integrinas/química , Magnésio/farmacologia , Manganês/farmacologia , Camundongos , NAD/farmacologia , Ligação Proteica/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos
5.
Exp Cell Res ; 298(1): 28-37, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15242759

RESUMO

We describe a novel interaction between the disintegrin and cysteine-rich (DC) domains of ADAM12 and the integrin alpha7beta1. Integrin alpha7beta1 extracted from human embryonic kidney 293 cells transfected with alpha7 cDNA was retained on an affinity column containing immobilized DC domain of ADAM12. 293 cells stably transfected with alpha7 cDNA adhered to DC-coated wells, and this adhesion was partially inhibited by 6A11 integrin alpha7 function-blocking antibody. The X1 and the X2 extracellular splice variants of integrin alpha7 supported equally well adhesion to the DC protein. Integrin alpha7beta1-mediated cell adhesion to DC had different requirements for Mn2+ than adhesion to laminin. Furthermore, integrin alpha7beta1-mediated cell adhesion to laminin, but not to DC, resulted in efficient cell spreading and phosphorylation of focal adhesion kinase (FAK) at Tyr397. We also show that adhesion of L6 myoblasts to DC is mediated in part by the endogenous integrin alpha7beta1 expressed in these cells. Since integrin alpha7 plays an important role in muscle cell growth, stability, and survival, and since ADAM12 has been implicated in muscle development and regeneration, we postulate that the interaction between ADAM12 and integrin alpha7beta1 may be relevant to muscle development, function, and disease. We also conclude that laminin and the DC domain of ADAM12 represent two functional ligands for integrin alpha7beta1, and adhesion to each of these two ligands via integrin alpha7beta1 triggers different cellular responses.


Assuntos
Adesão Celular/fisiologia , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Mioblastos/metabolismo , Proteínas ADAM , Proteína ADAM12 , Processamento Alternativo/genética , Animais , Sítios de Ligação/genética , Adesão Celular/genética , Linhagem Celular , Cisteína/metabolismo , Desintegrinas/metabolismo , Células Epiteliais/ultraestrutura , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Integrinas/genética , Laminina/metabolismo , Ligantes , Manganês/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mioblastos/ultraestrutura , Fosforilação , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Tirosina Quinases/metabolismo , Ratos , Transfecção , Tirosina/metabolismo
6.
J Biol Chem ; 279(30): 31197-204, 2004 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-15159400

RESUMO

Various lines of evidence suggest that communication between tropomyosin and myosin in the regulation of vertebrate-striated muscle contraction involves yet unknown changes in actin conformation. Possible participation of loop 38-52 in this communication has recently been questioned based on unimpaired Ca(2+) regulation of myosin interaction, in the presence of the tropomyosin-troponin complex, with actin cleaved by subtilisin between Met(47) and Gly(48). We have compared the effects of actin cleavage by subtilisin and by protease ECP32, between Gly(42) and Val(43), on its interaction with myosin S1 in the presence and absence of tropomyosin or tropomyosin-troponin. Both individual modifications reduced activation of S1 ATPase by actin to a similar extent. The effect of ECP cleavage, but not of subtilisin cleavage, was partially reversed by stabilization of interprotomer contacts with phalloidin, indicating different pathways of signal transmission from the N- and C-terminal parts of loop 38-52 to myosin binding sites. ECP cleavage diminished the affinity to tropomyosin and reduced its inhibition of acto-S1 ATPase at low S1 concentrations, but increased the tropomyosin-mediated cooperative enhancement of the ATPase by S1 binding to actin. These effects were reversed by phalloidin. Subtilisin-cleaved actin more closely resembled unmodified actin than the ECP-modified actin. Limited proteolysis of the modified and unmodified F-actins revealed an allosteric effect of ECP cleavage on the conformation of the actin subdomain 4 region that is presumably involved in tropomyosin binding. Our results point to a possible role of the N-terminal part of loop 38-52 of actin in communication between tropomyosin and myosin through changes in actin structure.


Assuntos
Actinas/química , Actinas/metabolismo , Desoxirribonuclease I/metabolismo , Miosinas/metabolismo , Tropomiosina/metabolismo , Troponina/metabolismo , Animais , Sítios de Ligação , Endopeptidases/metabolismo , Técnicas In Vitro , Cinética , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Subfragmentos de Miosina/metabolismo , Faloidina/farmacologia , Coelhos , Transdução de Sinais , Subtilisina/metabolismo
7.
Mol Cell Biol ; 23(19): 6725-38, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12972593

RESUMO

Skeletal myoblasts grown in vitro and induced to differentiate either form differentiated multinucleated myotubes or give rise to quiescent, undifferentiated "reserve cells" that share several characteristics with muscle satellite cells. The mechanism of determination of reserve cells is poorly understood. We find that the expression level of the metalloprotease disintegrin ADAM12 is much higher in proliferating C2C12 myoblasts and in reserve cells than in myotubes. Inhibition of ADAM12 expression in differentiating C2C12 cultures by small interfering RNA is accompanied by lower expression levels of both quiescence markers (retinoblastoma-related protein p130 and cell cycle inhibitor p27) and differentiation markers (myogenin and integrin alpha7A isoform). Overexpression of ADAM12 in C2C12 cells under conditions that promote cell cycle progression leads to upregulation of p130 and p27, cell cycle arrest, and downregulation of MyoD. Thus, enhanced expression of ADAM12 induces a quiescence-like phenotype and does not stimulate differentiation. We also show that the region extending from the disintegrin to the transmembrane domain of ADAM12 and containing cell adhesion activity as well as the cytoplasmic domain of ADAM12 are required for ADAM12-mediated cell cycle arrest, while the metalloprotease domain is not essential. Our results suggest that ADAM12-mediated adhesion and/or signaling may play a role in determination of the pool of reserve cells during myoblast differentiation.


Assuntos
Desintegrinas/metabolismo , Proteínas de Membrana/fisiologia , Metaloendopeptidases/metabolismo , Proteínas Musculares/fisiologia , Músculo Esquelético/embriologia , Proteínas ADAM , Proteína ADAM12 , Animais , Biomarcadores , Proteínas de Transporte/metabolismo , Ciclo Celular , Diferenciação Celular/fisiologia , Divisão Celular , Linhagem Celular , Regulação da Expressão Gênica , Cadeias alfa de Integrinas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Fibras Musculares Esqueléticas/fisiologia , Proteínas Musculares/química , Proteínas Musculares/genética , Mutação , Proteína MyoD/metabolismo , Miogenina/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Proteína 1 de Ligação ao Retinoblastoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA