Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 18736, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37907514

RESUMO

During atherosclerotic plaque formation, smooth muscle cells (SMCs) switch from a contractile/differentiated to a synthetic/dedifferentiated phenotype. We previously isolated differentiated spindle-shaped (S) and dedifferentiated rhomboid (R) SMCs from porcine coronary artery. R-SMCs express S100A4, a calcium-binding protein. We investigated the role of apelin in this phenotypic conversion, as well as its relationship with S100A4. We found that apelin was highly expressed in R-SMCs compared with S-SMCs. We observed a nuclear expression of apelin in SMCs within experimentally-induced intimal thickening of the porcine coronary artery and rat aorta. Plasmids targeting apelin to the nucleus (N. Ap) and to the secretory vesicles (S. Ap) were transfected into S-SMCs where apelin was barely detectable. Both plasmids induced the SMC transition towards a R-phenotype. Overexpression of N. Ap, and to a lesser degree S. Ap, led to a nuclear localization of S100A4. Stimulation of S-SMCs with platelet-derived growth factor-BB, known to induce the transition toward the R-phenotype, yielded the direct interaction and nuclear expression of both apelin and S100A4. In conclusion, apelin induces a SMC phenotypic transition towards the synthetic phenotype. These results suggest that apelin acts via nuclear re-localization of S100A4, raising the possibility of a new pro-atherogenic relationship between apelin and S100A4.


Assuntos
Aterosclerose , Animais , Ratos , Apelina/genética , Apelina/metabolismo , Aterosclerose/metabolismo , Movimento Celular , Células Cultivadas , Miócitos de Músculo Liso/metabolismo , Fenótipo , Suínos
2.
J Cell Physiol ; 237(3): 1980-1991, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34988986

RESUMO

Nifedipine, an L-type voltage-gated Ca2+ channel (L-VGCC) blocker, is one of the most used tocolytics to treat preterm labor. In clinical practice, nifedipine efficiently decreases uterine contractions, but its efficacy is limited over time, and repeated or maintained nifedipine-based tocolysis appears to be ineffective in preventing preterm birth. We aimed to understand why nifedipine has short-lasting efficiency for the inhibition of uterine contractions. We used ex vivo term pregnant human myometrial strips treated with cumulative doses of nifedipine. We observed that nifedipine inhibited spontaneous myometrial contractions in tissues with high and regular spontaneous contractions. By contrast, nifedipine appeared to increase contractions in tissues with low and/or irregular spontaneous contractions. To investigate the molecular mechanisms activated by nifedipine in myometrial cells, we used the pregnant human myometrial cell line PHM1-41 that does not express L-VGCC. The in vitro measurement of intracellular Ca2+ showed that high doses of nifedipine induced an important intracellular Ca2+ entry in myometrial cells. The inhibition or downregulation of the genes encoding for store-operated Ca2+ entry channels from the Orai and transient receptor potential-canonical (TRPC) families in PHM1-41 cells highlighted the implication of TRPC1 in nifedipine-induced Ca2+ entry. In addition, the use of 2-APB in combination with nifedipine on human myometrial strips tends to confirm that the pro-contractile effect induced by nifedipine on myometrial tissues may involve the activation of TRPC channels.


Assuntos
Contração Muscular , Miométrio , Nifedipino , Canais de Cátion TRPC , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Feminino , Humanos , Contração Muscular/efeitos dos fármacos , Miométrio/efeitos dos fármacos , Nifedipino/farmacologia , Gravidez , Nascimento Prematuro/metabolismo , Nascimento Prematuro/prevenção & controle , Canais de Cátion TRPC/metabolismo , Contração Uterina
3.
Mol Ther ; 24(9): 1675-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27377043

RESUMO

Cell penetrating peptides (CPPs) from the protein ZEBRA are promising candidates to exploit in therapeutic cancer vaccines, since they can transport antigenic cargos into dendritic cells and induce tumor-specific T cells. Employing CPPs for a given cancer indication will require engineering to include relevant tumor-associated epitopes, administration with an appropriate adjuvant, and testing for antitumor immunity. We assessed the importance of structural characteristics, efficiency of in vitro transduction of target cells, and choice of adjuvant in inducing the two key elements in antitumor immunity, CD4 and CD8 T cells, as well as control of tumor growth in vivo. Structural characteristics associated with CPP function varied according to CPP truncations and cargo epitope composition, and correlated with in vitro transduction efficiency. However, subsequent in vivo capacity to induce CD4 and CD8 T cells was not always predicted by in vitro results. We determined that the critical parameter for in vivo efficacy using aggressive mouse tumor models was the choice of adjuvant. Optimal pairing of a particular ZEBRA-CPP sequence and antigenic cargo together with adjuvant induced potent antitumor immunity. Our results highlight the irreplaceable role of in vivo testing of novel vaccine constructs together with adjuvants to select combinations for further development.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Peptídeos Penetradores de Células/imunologia , Neoplasias/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Dicroísmo Circular , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Camundongos , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Transativadores/química , Transativadores/imunologia , Resultado do Tratamento , Vacinação
4.
Cell Calcium ; 56(6): 513-21, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25468730

RESUMO

Cytosolic Ca(2+) signals are fundamental for the early and late steps of myoblast differentiation and are, as in many cells, generated by Ca(2+) release from internal stores as well as by plasma membrane Ca(2+) entry. Our recent studies identified the store-operated Ca(2+) channels, Orai1 and TRPC1&C4, as crucial for the early steps of human myogenesis and for the late fusion events. In the present work, we assessed the role of the inositol-1,4,5 tris-phosphate receptor (IP3R) type 1 during human myoblast differentiation. We demonstrated, using siRNA strategy that IP3R1 is required for the expression of muscle-specific transcription factors such as myogenin and MEF2 (myocyte enhancer factor 2), and for the formation of myotubes. The knockdown of IP3R1 strongly reduced endogenous spontaneous Ca(2+) transients, and attenuated store-operated Ca(2+) entry. As well, two Ca(2+)-dependent key enzymes of muscle differentiation, NFAT and CamKII are down-regulated upon siIP3R1 treatment. On the contrary, the overexpression of IP3R1 accelerated myoblasts differentiation. These findings identify Ca(2+) release mediated by IP3R1 as an essential mechanism during the early steps of myoblast differentiation.


Assuntos
Diferenciação Celular/fisiologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/fisiologia , Cálcio/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/genética , Fatores de Transcrição MEF2/fisiologia , Miogenina/fisiologia , Fatores de Transcrição NFATC/fisiologia , RNA Interferente Pequeno/farmacologia
5.
Curr Biol ; 24(15): 1679-88, 2014 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-25042585

RESUMO

BACKGROUND: Excitatory synapses in the CNS are highly dynamic structures that can show activity-dependent remodeling and stabilization in response to learning and memory. Synapses are enveloped with intricate processes of astrocytes known as perisynaptic astrocytic processes (PAPs). PAPs are motile structures displaying rapid actin-dependent movements and are characterized by Ca(2+) elevations in response to neuronal activity. Despite a debated implication in synaptic plasticity, the role of both Ca(2+) events in astrocytes and PAP morphological dynamics remain unclear. RESULTS: In the hippocampus, we found that PAPs show extensive structural plasticity that is regulated by synaptic activity through astrocytic metabotropic glutamate receptors and intracellular calcium signaling. Synaptic activation that induces long-term potentiation caused a transient PAP motility increase leading to an enhanced astrocytic coverage of the synapse. Selective activation of calcium signals in individual PAPs using exogenous metabotropic receptor expression and two-photon uncaging reproduced these effects and enhanced spine stability. In vivo imaging in the somatosensory cortex of adult mice revealed that increased neuronal activity through whisker stimulation similarly elevates PAP movement. This in vivo PAP motility correlated with spine coverage and was predictive of spine stability. CONCLUSIONS: This study identifies a novel bidirectional interaction between synapses and astrocytes, in which synaptic activity and synaptic potentiation regulate PAP structural plasticity, which in turn determines the fate of the synapse. This mechanism may represent an important contribution of astrocytes to learning and memory processes.


Assuntos
Astrócitos/metabolismo , Potenciação de Longa Duração , Plasticidade Neuronal , Sinapses/fisiologia , Animais , Sinalização do Cálcio , Feminino , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Glutamato Metabotrópico/metabolismo , Córtex Somatossensorial/fisiologia , Imagem com Lapso de Tempo
6.
PLoS One ; 8(8): e71770, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967242

RESUMO

Initiation of human myoblast differentiation requires a negative shift (hyperpolarization) of the resting potential of myoblasts that depends on the activation of Kir2.1 potassium channels. These channels are regulated by a tyrosine phosphorylation. Using human primary myoblast culture, we investigated a possible role of various receptor tyrosine kinases in the induction of the differentiation process. We found that Epidermal Growth Factor Receptor (EGFR) is a key regulator of myoblast differentiation. EGFR activity is down-regulated during early human myoblast differentiation, and this event is required for normal differentiation to take place. Furthermore, EGFR silencing in proliferation conditions was able to trigger the differentiation program. This occurs through an increase of Kir2.1 channel activity that, via a rise of store-operated Ca(2+) entry, leads to the expression of myogenic transcription factors and muscle specific proteins (Myogenin, Myocyte Enhancer Factor 2 (MEF2), Myosin Heavy Chain (MyHC)). Finally, blocking myoblast cell cycle in proliferation conditions using a cdk4 inhibitor greatly decreased myoblast proliferation but was not able, on its own, to promote myoblast differentiation. Taken together, these results show that EGFR down-regulation is an early event that is required for the induction of myoblast differentiation.


Assuntos
Diferenciação Celular/genética , Receptores ErbB/genética , Mioblastos/citologia , Mioblastos/metabolismo , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ativação Transcricional
7.
J Cell Biol ; 194(2): 335-46, 2011 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-21788372

RESUMO

Cytosolic Ca(2+) signals encoded by repetitive Ca(2+) releases rely on two processes to refill Ca(2+) stores: Ca(2+) reuptake from the cytosol and activation of a Ca(2+) influx via store-operated Ca(2+) entry (SOCE). However, SOCE activation is a slow process. It is delayed by >30 s after store depletion because stromal interaction molecule 1 (STIM1), the Ca(2+) sensor of the intracellular stores, must form clusters and migrate to the membrane before being able to open Orai1, the plasma membrane Ca(2+) channel. In this paper, we identify a new protein, STIM1L, that colocalizes with Orai1 Ca(2+) channels and interacts with actin to form permanent clusters. This property allowed the immediate activation of SOCE, a characteristic required for generating repetitive Ca(2+) signals with frequencies within seconds such as those frequently observed in excitable cells. STIM1L was expressed in several mammalian tissues, suggesting that many cell types rely on this Ca(2+) sensor for their Ca(2+) homeostasis and intracellular signaling.


Assuntos
Actinas/metabolismo , Processamento Alternativo/genética , Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Canais de Cálcio/metabolismo , Células Cultivadas , Éxons/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos , Proteínas de Neoplasias/genética , Proteína ORAI1 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Molécula 1 de Interação Estromal
8.
Cell Calcium ; 49(2): 115-27, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21193229

RESUMO

The ER Ca²+ sensor STIM1 and the Ca²+ channel Orai1 are key players in store-operated Ca²+ entry (SOCE). In addition, channels from the TRPC family were also shown to be engaged during SOCE, while their precise implication remains controversial. In this study, we investigated the molecular players involved in SOCE triggered by the SERCA pump inhibitor thapsigargin in an endothelial cell line, the EA.hy926. siRNA directed against STIM1 or Orai1 reduced Ca²+ entry by about 50-60%, showing that a large part of the entry is independent from these proteins. Blocking the PLC or the PKC pathway completely abolished thapsigargin-induced Ca²+ entry in cells depleted from STIM1 and/or Orai1. The phorbol ester PMA or the DAG analog OAG restored the Ca²+ entry inhibited by PLC blockers, showing an involvement of PLC/PKC pathway in SOCE. Using pharmacological inhibitors or siRNA revealed that the PKCeta is required for Ca²+ entry, and pharmacological inhibition of the tyrosine kinase Src also reduced Ca²+ entry. TRPC3 silencing diminished the entry by 45%, while the double STIM1/TRPC3 invalidation reduced Ca²+ entry by more than 85%. Hence, in EA.hy926 cells, TG-induced Ca²+ entry results from the activation of the STIM1/Orai1 machinery, and from the activation of TRPC3.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Células Endoteliais/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Tapsigargina/farmacologia , Canais de Cálcio/genética , Linhagem Celular , Células Endoteliais/enzimologia , Fura-2/farmacologia , Humanos , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Proteína ORAI1 , Ésteres de Forbol/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
9.
J Biol Chem ; 285(29): 22437-47, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20436167

RESUMO

Our recent work identified store-operated Ca(2+) entry (SOCE) as the critical Ca(2+) source required for the induction of human myoblast differentiation (Darbellay, B., Arnaudeau, S., König, S., Jousset, H., Bader, C., Demaurex, N., and Bernheim, L. (2009) J. Biol. Chem. 284, 5370-5380). The present work indicates that STIM2 silencing, similar to STIM1 silencing, reduces myoblast SOCE amplitude and differentiation. Because myoblasts in culture can be induced to differentiate into myotubes, which spontaneously contract in culture, we used the same molecular tools to explore whether the Ca(2+) mechanism of excitation-contraction coupling also relies on STIM1 and STIM2. Live cell imaging of early differentiating myoblasts revealed a characteristic clustering of activated STIM1 and STIM2 during the first few hours of differentiation. Thapsigargin-induced depletion of endoplasmic reticulum Ca(2+) content caused STIM1 and STIM2 redistribution into clusters, and co-localization of both STIM proteins. Interaction of STIM1 and STIM2 was revealed by a rapid increase in fluorescence resonance energy transfer between CFP-STIM1 and YFP-STIM2 after SOCE activation and confirmed by co-immunoprecipitation of endogenous STIM1 and STIM2. Although both STIM proteins clearly contribute to SOCE and are required during the differentiation process, STIM1 and STIM2 are functionally largely redundant as overexpression of either STIM1 or STIM2 corrected most of the impact of STIM2 or STIM1 silencing on SOCE and differentiation. With respect to excitation-contraction, we observed that human myotubes rely also on STIM1 and STIM2 to refill their endoplasmic reticulum Ca(2+)-content during repeated KCl-induced Ca(2+) releases. This indicates that STIM2 is a necessary partner of STIM1 for excitation-contraction coupling. Thus, both STIM proteins are required and interact to control SOCE during human myoblast differentiation and human myotube excitation-contraction coupling.


Assuntos
Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Acoplamento Excitação-Contração , Proteínas de Membrana/metabolismo , Músculos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Pré-Escolar , Inativação Gênica , Humanos , Potenciais da Membrana , Fibras Musculares Esqueléticas/metabolismo , Músculos/citologia , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal , Regulação para Cima
10.
J Biol Chem ; 284(8): 5370-80, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19088073

RESUMO

Our previous work on human myoblasts suggested that a hyperpolarization followed by a rise in [Ca(2+)](in) involving store-operated Ca(2+) entry (SOCE) channels induced myoblast differentiation. Advances in the understanding of the SOCE pathway led us to examine more precisely its role in post-natal human myoblast differentiation. We found that SOCE orchestrated by STIM1, the endoplasmic reticulum Ca(2+) sensor activating Orai Ca(2+) channels, is crucial. Silencing STIM1, Orai1, or Orai3 reduced SOCE amplitude and myoblast differentiation, whereas Orai2 knockdown had no effect. Conversely, overexpression of STIM1 with Orai1 increased SOCE and accelerated myoblast differentiation. STIM1 or Orai1 silencing decreased resting [Ca(2+)](in) and intracellular Ca(2+) store content, but correction of these parameters did not rescue myoblast differentiation. Remarkably, SOCE amplitude correlated linearly with the expression of two early markers of myoblast differentiation, MEF2 and myogenin, regardless of the STIM or Orai isoform that was silenced. Unexpectedly, we found that the hyperpolarization also depends on SOCE, placing SOCE upstream of K(+) channel activation in the signaling cascade that controls myoblast differentiation. These findings indicate that STIM1 and Orai1 are key molecules for the induction of human myoblast differentiation.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Diferenciação Celular/fisiologia , Potenciais da Membrana/fisiologia , Proteínas de Membrana/metabolismo , Mioblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Canais de Cálcio/genética , Células Cultivadas , Pré-Escolar , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lactente , Masculino , Proteínas de Membrana/genética , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Miogenina/genética , Miogenina/metabolismo , Proteínas de Neoplasias/genética , Proteína ORAI1 , Molécula 1 de Interação Estromal
11.
Development ; 135(5): 859-67, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18216177

RESUMO

Myoblast differentiation is essential to skeletal muscle formation and repair. The earliest detectable event leading to human myoblast differentiation is an upregulation of Kir2.1 channel activity, which causes a negative shift (hyperpolarization) of the resting potential of myoblasts. After exploring various mechanisms, we found that this upregulation of Kir2.1 was due to dephosphorylation of the channel itself. Application of genistein, a tyrosine kinase inhibitor, increased Kir2.1 activity and triggered the differentiation process, whereas application of bpV(Phen), a tyrosine phosphatase inhibitor, had the opposite effects. We could show that increased Kir2.1 activity requires dephosphorylation of tyrosine 242; replacing this tyrosine in Kir2.1 by a phenylalanine abolished inhibition by bpV(Phen). Finally, we found that the level of tyrosine phosphorylation in endogenous Kir2.1 channels is considerably reduced during differentiation when compared with proliferation. We propose that Kir2.1 channels are already present at the membrane of proliferating, undifferentiated human myoblasts but in a silent state, and that Kir2.1 tyrosine 242 dephosphorylation triggers differentiation.


Assuntos
Diferenciação Celular , Mioblastos/citologia , Mioblastos/fisiologia , Fosfotirosina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Tirosina , Sequência de Aminoácidos , Biotinilação , Técnicas de Cultura de Células , Divisão Celular , Membrana Celular/fisiologia , DNA/genética , Eletrofisiologia , Eletroporação , Regulação da Expressão Gênica , Humanos , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Transfecção , Tirosina/metabolismo , Regulação para Cima
12.
Mol Ther ; 16(2): 404-10, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18026170

RESUMO

Cell therapy for Duchenne muscular dystrophy and other muscle diseases is limited by a massive early cell death following injections. In this study, we explored the potential benefit of heme oxygenase-1 (HO-1) expression in the survival of porcine myogenic precursor cells (MPCs) transplanted in pig skeletal muscle. Increased HO-1 expression was assessed either by transient hyperthermia or by HO-1 lentiviral infection. One day after the thermic shock, we observed a fourfold and a threefold increase in HSP70/72 and HO-1 levels, respectively. This treatment protected 30% of cells from staurosporine-induced apoptosis in vitro. When porcine MPC were heat-shocked prior to grafting, we improved cell survival by threefold at 5 days after autologous transplantation (26.3 +/- 5.5% surviving cells). After HO-1 lentiviral transduction, almost 60% of cells expressed the transgene and kept their myogenic properties to proliferate and fuse in vitro. Apoptosis of HO-1 transduced cells was reduced by 50% in vitro after staurosporine induction. Finally, a fivefold enhancement in cell survival was observed after transplantation of HO-1-group (47.5 +/- 9.1% surviving cells) as compared to the nls-LacZ-group or control group. These results identify HO-1 as a protective gene against early MPC death post-transplantation.


Assuntos
Heme Oxigenase-1/metabolismo , Lentivirus/genética , Mioblastos/citologia , Mioblastos/transplante , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Citometria de Fluxo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Heme Oxigenase-1/genética , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/metabolismo , RNA Interferente Pequeno/genética , Estaurosporina/farmacologia , Suínos , Transplante Autólogo
13.
Development ; 133(16): 3107-14, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16831831

RESUMO

In human myoblasts triggered to differentiate, a hyperpolarization, resulting from K+ channel (Kir2.1) activation, allows the generation of an intracellular Ca2+ signal. This signal induces an increase in expression/activity of two key transcription factors of the differentiation process, myogenin and MEF2. Blocking hyperpolarization inhibits myoblast differentiation. The link between hyperpolarization-induced Ca2+ signals and the four main regulatory pathways involved in myoblast differentiation was the object of this study. Of the calcineurin, p38-MAPK, PI3K and CaMK pathways, only the calcineurin pathway was inhibited when Kir2.1-linked hyperpolarization was blocked. The CaMK pathway, although Ca2+ dependent, is unaffected by changes in membrane potential or block of Kir2.1 channels. Concerning the p38-MAPK and PI3K pathways, their activity is present already in proliferating myoblasts and they are unaffected by hyperpolarization or Kir2.1 channel block. We conclude that the Kir2.1-induced hyperpolarization triggers human myoblast differentiation via the activation of the calcineurin pathway, which, in turn, induces expression/activity of myogenin and MEF2.


Assuntos
Calcineurina/metabolismo , Sinalização do Cálcio , Diferenciação Celular , Mioblastos/citologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fusão Celular , Membrana Celular/metabolismo , Polaridade Celular , Humanos , Fatores de Transcrição MEF2 , Mioblastos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Miogenina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Cell Physiol ; 208(2): 435-45, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16688780

RESUMO

Increases in cytoplasmic Ca(2+) are crucial for inducing the initial steps of myoblast differentiation that ultimately lead to fusion; yet the mechanisms that produce this elevated Ca(2+) have not been fully resolved. For example, it is still unclear whether the increase comes exclusively from membrane Ca(2+) influx or also from Ca(2+) release from internal stores. To address this, we investigated early differentiation of myoblast clones each derived from single post-natal human satellite cells. Initial differentiation was assayed by immunostaining myonuclei for the transcription factor MEF2. When Ca(2+) influx was eliminated by using low external Ca(2+) media, we found that approximately half the clones could still differentiate. Of the clones that required influx of external Ca(2+), most clones used T-type Ca(2+) channels, but others used store-operated channels as influx-generating mechanisms. On the other hand, clones that differentiated in low external Ca(2+) relied on Ca(2+) release from internal stores through IP(3) receptors. Interestingly, by following clones over time, we observed that some switched their preferred Ca(2+) source: clones that initially used calcium release from internal stores to differentiate later required Ca(2+) influx and inversely. In conclusion, we show that human myoblasts can use three alternative mechanisms to increase cytoplasmic Ca(2+) at the onset of the differentiation process: influx through T-types Ca(2+) channels, influx through store operated channels and release from internal stores through IP(3) receptors. In addition, we suggest that, probably because Ca(2+) elevation is essential during initial differentiation, myoblasts may be able to select between these alternate Ca(2+) pathways.


Assuntos
Cálcio/metabolismo , Diferenciação Celular , Mioblastos/citologia , Mioblastos/fisiologia , Canais de Cálcio/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Criança , Pré-Escolar , Células Clonais , Humanos , Imuno-Histoquímica , Lactente , Modelos Biológicos , Mioblastos/efeitos dos fármacos , Tapsigargina/farmacologia
15.
Neuromuscul Disord ; 15(3): 237-44, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15725585

RESUMO

Myoblast transplantation is a potential therapy for severe muscle trauma, myopathies and heart infarct. Success with this therapy relies on the ability to obtain cell preparations enriched in myogenic precursor cells and on their survival after transplantation. To define myoblast transplantation strategies applicable to patients, we used a large animal model, the pig. Muscle dissociation procedures adapted to porcine tissue gave high yields of cells containing at least 80% myogenic precursor cells. Autologous transplantation of 3[H]-thymidine labeled porcine myogenic precursor cells indicated 60% survival at day 1 followed by a decay to 10% at day 5 post-injection. Nuclei of myogenic precursor cells transduced with a lentivirus encoding the nls-lacZ reporter gene were present in host myotubes 8 days post-transplantation, indicating that injected myogenic precursor cells contribute to muscle regeneration. This work suggests that pig is an adequate large animal model for exploring myogenic precursor cells transplantation strategies applicable in patients.


Assuntos
Músculo Esquelético/transplante , Mioblastos/transplante , Transplante Autólogo , Animais , Antígeno CD56/metabolismo , Contagem de Células/métodos , Proliferação de Células , Sobrevivência Celular/fisiologia , Desmina/metabolismo , Feminino , Citometria de Fluxo , Indóis , Desenvolvimento Muscular , Músculo Esquelético/fisiologia , Mioblastos/fisiologia , Suínos , Timidina/metabolismo , Fatores de Tempo , Transdução Genética , Trítio/metabolismo
16.
Circ Res ; 95(12): e98-109, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15539631

RESUMO

ANP (atrial natriuretic peptide) is widely recognized as an important vasorelaxant, diuretic, and cardioprotective hormone. Little is known, however, about how ANP-secretory vesicles form within the atrial myocytes. Secretory vesicles were visualized by fluorescence microscope imaging in live rat atrial myocytes expressing proANP-enhanced green fluorescent protein (EGFP), or N-terminal-mutated fusion proteins thought to suppress the calcium-dependent aggregation of proANP. Results showed the following: (1) aggregates of proANP and coexpressed proANP-EGFP recruited peptidylglycine alpha-amidating monooxygenase (PAM)-1, an abundant atrial integral vesicle membrane protein; (2) coexpressed N-terminal-mutated (Glu23,24-->Gln23,24) and N-terminal-deleted proANP-EGFP inhibited recruitment of PAM-1 by up to 60%; (3) 4-phenyl-3-butenoic acid (PBA) (10 mumol/L), a pharmacological inhibitor of the lumenal peptidylglycine alpha-hydroxylating monooxygenase domain of PAM proteins, inhibited recruitment of endogenous PAM-1 and of coexpressed pro-EGFP-PAM-1; (4) PBA had no effect on exocytosis of the potassium inward rectifier KIR2.1; (5) PBA induced a deformation of the secretory vesicles but did not inhibit docking. These findings suggest that recruitment of PAM-1 to secretory vesicles depends on intact N-terminal proANP and on the lumenal domain of PAM-1. Conversely, PAM-1 participates in shaping the proANP-secretory vesicles. The full text of this article is available online at http://circres.ahajournals.org.


Assuntos
Apêndice Atrial/metabolismo , Fator Natriurético Atrial/fisiologia , Ácidos Graxos Monoinsaturados/farmacologia , Oxigenases de Função Mista/fisiologia , Complexos Multienzimáticos/fisiologia , Vesículas Secretórias/fisiologia , Animais , Apêndice Atrial/ultraestrutura , Sítios de Ligação , Eletroporação , Exocitose/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Oxigenases de Função Mista/antagonistas & inibidores , Complexos Multienzimáticos/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/fisiologia , Vesículas Secretórias/enzimologia , Vesículas Secretórias/ultraestrutura , Deleção de Sequência , Relação Estrutura-Atividade
17.
J Biol Chem ; 279(27): 28187-96, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15084602

RESUMO

It is widely thought that myogenin is one of the earliest detectable markers of skeletal muscle differentiation. Here we show that, during human myoblast differentiation, an inward rectifier K(+) channel (Kir2.1) and its associated hyperpolarization trigger expression and activity of the myogenic transcription factors, myogenin and myocyte enhancer factor-2 (MEF2). Furthermore, Kir2.1 current precedes and is required for the developmental increase in expression/activity of myogenin and MEF2. Drugs or antisense reducing Kir2.1 current diminished or suppressed fusion as well as expression/activity of myogenin and MEF2. In contrast, LY294002, an inhibitor of phosphatidylinositol 3-kinase (a pathway controlling initiation of the myogenic program) that inhibited both myogenin/MEF2 expression and fusion, did not affect Kir2.1 current. This non-blockade by LY294002 indicates that Kir2.1 acts upstream of myogenin and MEF2. We propose that Kir2.1 channel activation is a required key early event that initiates myogenesis by turning on myogenin and MEF2 transcription factors via a hyperpolarization-activated Ca(2+)-dependent pathway.


Assuntos
Membrana Celular/metabolismo , Proteínas de Ligação a DNA/biossíntese , Mioblastos/metabolismo , Miogenina/biossíntese , Fatores de Transcrição/biossíntese , Western Blotting , Cálcio/metabolismo , Calibragem , Diferenciação Celular , Divisão Celular , Células Cultivadas , Cromonas/farmacologia , Eletrofisiologia , Genes Reporter , Humanos , Imuno-Histoquímica , Íons , Cinética , Fatores de Transcrição MEF2 , Potenciais da Membrana , Microscopia Confocal , Microscopia de Fluorescência , Morfolinas/farmacologia , Fatores de Regulação Miogênica , Oligonucleotídeos Antissenso/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção
18.
Hum Gene Ther ; 14(16): 1525-33, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14577914

RESUMO

Conditionally immortalized human cells are valuable substrates for basic biologic studies, as well as for the production of specific proteins and for the creation of bioartificial organs. We previously demonstrated that the lentivector-mediated transduction of immortalizing genes into human primary cells is an efficient method for obtaining such cell lines. Here, we used human muscle satellite cells as model targets to examine the impact of the transduced genes on the genotypic and phenotypic characteristics of the immortalized cells. The most commonly used immortalizing gene, the SV40 large T antigen (T-Ag), was extremely efficient at inducing the continuous growth of primary myoblasts, but the resulting cells rapidly accumulated major chromosomal aberrations and exhibited profound phenotypic changes. In contrast, the constitutive expression of telomerase and Bmi-1 in satellite cells from a control individual and from a patient suffering from Duchenne's muscular dystrophy yielded cell lines that remained diploid and conserved their growth factor dependence for proliferation. However, despite the absence of detectable cytogenetic abnormalities, clones derived from satellite cells of a control individual exhibited a differentiation block in vitro. In contrast, a Duchenne-derived cell line exhibited all the phenotypic characteristics of its primary parent, including an ability to differentiate fully into myotubes when placed in proper culture conditions. This cell line should constitute a useful reagent for a wide range of studies aimed at this disease.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Linhagem Celular , Vetores Genéticos , Lentivirus/genética , Mioblastos , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Células Satélites de Músculo Esquelético/metabolismo , Telomerase/genética , Adolescente , Diferenciação Celular , Divisão Celular , Transplante de Células , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Cariotipagem , Masculino , Distrofia Muscular de Duchenne/patologia , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Telomerase/metabolismo
19.
Development ; 130(15): 3437-46, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12810591

RESUMO

We have previously shown that human myoblasts do not fuse when their voltage fails to reach the domain of a window T-type Ca(2+) current. We demonstrate, by changing the voltage in the window domain, that the Ca(2+) signal initiating fusion is not of the all-or-none type, but can be graded and is interpreted as such by the differentiation program. This was carried out by exploiting the properties of human ether-à-go-go related gene K(+) channels that we found to be expressed in human myoblasts. Methanesulfonanilide class III antiarrhythmic agents or antisense-RNA vectors were used to suppress completely ether-à-go-go related gene current. Both procedures induced a reproducible depolarization from -74 to -64 mV, precisely in the window domain where the T-type Ca(2+) current increases with voltage. This 10 mV depolarization raised the cytoplasmic free Ca(2+) concentration, and triggered a tenfold acceleration of myoblast fusion. Our results suggest that any mechanism able to modulate intracellular Ca(2+) concentration could affect the rate of myoblast fusion.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Mioblastos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Transativadores , Fusão Celular , Citoplasma/fisiologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Humanos , Potenciais da Membrana/fisiologia , Desenvolvimento Muscular/fisiologia , Canais de Potássio/metabolismo , Regulador Transcricional ERG
20.
Ann Thorac Surg ; 73(6): 1939-46, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12078794

RESUMO

BACKGROUND: The available techniques for intravascular gene delivery to the heart are inefficient and not organ-specific. Yet, effective treatment of heart failure will likely require transgene expression by the majority of cardiac myocytes. To address this problem, we developed a novel cannulation technique that achieves efficient isolation of the heart in situ using separate cardiopulmonary bypass (CPB) circuits for the heart and body in dogs. METHODS: The arterial inflow and venous effluent from the two circuits were physically isolated. The efficiency of separation was 98% to 99% in three preliminary experiments using Evans Blue dye-labeled albumin. In 6 dogs, the cardiac circuit was perfused with oxygenated crystalloid cardioplegia at 37 degrees C containing approximately 4 x 10(11) particles of an adenovirus encoding LacZ (AdCMVLacZ) with a perfusion pressure of 170 to 200 mm Hg for 15 minutes allowing virus to recirculate through the heart approximately 15 times. Cross-clamp time was 26 +/- 2 minutes and CPB time was 90 +/- 3 minutes. RESULTS: Five animals survived and were euthanized at 7 days. Beta-galactosidase activities measured using a chemiluminescent assay were three orders of magnitude higher in all areas of the heart than in the liver. Histological analyses revealed heterogeneous X-Gal staining of myocytes in all areas of the myocardium. CONCLUSIONS: Despite using a constitutive promoter, this technique yields relatively cardiac-specific transgene expression and is potentially translatable to clinical applications. Future studies will allow for further optimization of the conditions necessary for vector-mediated gene delivery to the heart.


Assuntos
Ponte Cardiopulmonar/métodos , Miocárdio , Transgenes , Adenoviridae/genética , Albuminas , Animais , Cães , Técnicas de Transferência de Genes , Coração/diagnóstico por imagem , Óperon Lac/genética , Miocárdio/patologia , Cintilografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...