Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 80, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36604419

RESUMO

Fibro-adipogenic progenitors (FAPs) play a crucial role in skeletal muscle regeneration, as they generate a favorable niche that allows satellite cells to perform efficient muscle regeneration. After muscle injury, FAP content increases rapidly within the injured muscle, the origin of which has been attributed to their proliferation within the muscle itself. However, recent single-cell RNAseq approaches have revealed phenotype and functional heterogeneity in FAPs, raising the question of how this differentiation of regenerative subtypes occurs. Here we report that FAP-like cells residing in subcutaneous adipose tissue (ScAT), the adipose stromal cells (ASCs), are rapidly released from ScAT in response to muscle injury. Additionally, we find that released ASCs infiltrate the damaged muscle, via a platelet-dependent mechanism and thus contribute to the FAP heterogeneity. Moreover, we show that either blocking ASCs infiltration or removing ASCs tissue source impair muscle regeneration. Collectively, our data reveal that ScAT is an unsuspected physiological reservoir of regenerative cells that support skeletal muscle regeneration, underlining a beneficial relationship between muscle and fat.


Assuntos
Músculo Esquelético , Doenças Musculares , Humanos , Tecido Adiposo , Diferenciação Celular/genética , Adipogenia/genética
2.
Front Cell Dev Biol ; 8: 598520, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33490065

RESUMO

To coordinate specialized organs, inter-tissue communication appeared during evolution. Consequently, individual organs communicate their states via a vast interorgan communication network (ICN) made up of peptides, proteins, and metabolites that act between organs to coordinate cellular processes under homeostasis and stress. However, the nature of the interorgan signaling could be even more complex and involve mobilization mechanisms of unconventional cells that are still poorly described. Mesenchymal stem/stromal cells (MSCs) virtually reside in all tissues, though the biggest reservoir discovered so far is adipose tissue where they are named adipose stromal cells (ASCs). MSCs are thought to participate in tissue maintenance and repair since the administration of exogenous MSCs is well known to exert beneficial effects under several pathological conditions. However, the role of endogenous MSCs is barely understood. Though largely debated, the presence of circulating endogenous MSCs has been reported in multiple pathophysiological conditions, but the significance of such cell circulation is not known and therapeutically untapped. In this review, we discuss current knowledge on the circulation of native MSCs, and we highlight recent findings describing MSCs as putative key components of the ICN.

3.
Int J Mol Sci ; 20(15)2019 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-31344837

RESUMO

The Rho GTPase family can be classified into classic and atypical members. Classic members cycle between an inactive Guanosine DiPhosphate -bound state and an active Guanosine TriPhosphate-bound state. Atypical Rho GTPases, such as RND1, are predominantly in an active GTP-bound conformation. The role of classic members in oncogenesis has been the subject of numerous studies, while that of atypical members has been less explored. Besides the roles of RND1 in healthy tissues, recent data suggest that RND1 is involved in oncogenesis and response to cancer therapeutics. Here, we present the current knowledge on RND1 expression, subcellular localization, and functions in healthy tissues. Then, we review data showing that RND1 expression is dysregulated in tumors, the molecular mechanisms involved in this deregulation, and the role of RND1 in oncogenesis. For several aggressive tumors, RND1 presents the features of a tumor suppressor gene. In these tumors, low expression of RND1 is associated with a bad prognosis for the patients. Finally, we highlight that RND1 expression is induced by anticancer agents and modulates their response. Of note, RND1 mRNA levels in tumors could be used as a predictive marker of both patient prognosis and response to anticancer agents.


Assuntos
Carcinogênese/genética , Neoplasias/genética , Proteínas rho de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes Supressores de Tumor , Humanos , Neoplasias/patologia
4.
Mol Cancer Res ; 17(2): 384-397, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30266751

RESUMO

Glioblastomas (GB) are malignant brain tumors with poor prognosis despite treatment with surgery and radio/chemotherapy. These tumors are defined by an important cellular heterogeneity and notably contain a subpopulation of GB-initiating cells (GIC), which contribute to tumor aggressiveness, resistance, and recurrence. Some integrins are specifically expressed by GICs and could be actionable targets to improve GB treatment. Here, integrin ß8 (ITGB8) was identified as a potential selective target in this highly tumorigenic GIC subpopulation. Using several patient-derived primocultures, it was demonstrated that ITGB8 is overexpressed in GICs compared with their differentiated progeny. Furthermore, ITGB8 is also overexpressed in GB, and its overexpression is correlated with poor prognosis and with the expression of several other classic stem cell markers. Moreover, inhibiting ITGB8 diminished several main GIC characteristics and features, including self-renewal ability, stemness, migration potential, and tumor formation capacity. Blockade of ITGB8 significantly impaired GIC cell viability via apoptosis induction. Finally, the combination of radiotherapy and ITGB8 targeting radiosensitized GICs through postmitotic cell death. IMPLICATIONS: This study identifies ITGB8 as a new selective marker for GICs and as a promising therapeutic target in combination with chemo/radiotherapy for the treatment of highly aggressive brain tumors.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Cadeias beta de Integrinas/metabolismo , Radiossensibilizantes/uso terapêutico , Animais , Diferenciação Celular , Humanos , Camundongos , Camundongos Nus , Radiossensibilizantes/farmacologia , Transfecção
5.
Oncotarget ; 9(73): 33788-33803, 2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30333910

RESUMO

Despite post-operative radio-chemotherapy, glioblastoma systematically locally recurs. Tumors contacting the periventricular zone (PVZ) show earlier and more distant relapses than tumors not contacting the PVZ. Since glioblastoma stem-like cells (GSCs) have been proposed to play a major role in glioblastoma recurrence, we decided to test whether GSC migration properties could be different according to their anatomical location (PVZ+/PVZ-). For that purpose, we established paired cultures of GSCs from the cortical area (CT) and the PVZ of glioblastoma patient tumors. We demonstrated that PVZ GSCs possess higher migration and invasion capacities than CT GSCs. We highlighted specific transcriptomic profiles in PVZ versus CT populations and identified a down-regulation of the RhoGTPase, RND1 in PVZ GSCs compared to CT GSCs. Overexpression of RND1, dramatically inhibited PVZ GSC migration and conversely, downregulation of RND1 increased CT GSC migration. Additionally, transcriptomic analyses also revealed a down-regulation of RND1 in glioblastoma compared to normal brain. Using the glioblastoma TCGA database, low levels of RND1 were also shown to correlate with a decreased overall survival of patients. Finally, based on signaling pathways activated in patients with low levels of RND1, we identified an RND1 low signature of six genes (MET, LAMC1, ITGA5, COL5A1, COL3A1, COL1A2) that is an independent prognostic factor in glioblastoma. These findings contribute to explain the shorter time to progression of patients with PVZ involvement and, point out genes that establish the RND1 low signature as key targets genes to impede tumor relapse after treatment.

6.
Cell Death Dis ; 9(9): 931, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30209297

RESUMO

RHO GTPases regulate essential functions such as the organization of the actin cytoskeleton. The classic members cycle between an active GTP-bound and an inactive GDP-bound conformation whereas atypical members are predominantly GTP-bound. Besides their well-established role, the classic RHO GTPases RHOB and RAC1, are rapidly induced and/or activated by genotoxic stress and contribute to the DNA damage response. Here we used camptothecin, a selective topoisomerase I (TOP1) inhibitor that stabilizes TOP1 cleavage complexes (TOP1cc), to search for other potential early DNA damage-inducible RHO GTPase genes. We identified that an atypical RHO GTPase, RND1, is rapidly induced by camptothecin. RND1 induction is closely associated with the presence of TOP1cc induced by camptothecin or by DNA lesions that elevate TOP1cc levels such as UV and hydrogen peroxide. We further demonstrated that camptothecin increases RND1 gene transcription and mRNA stability. Camptothecin also increases poly(ADP-ribose) polymerase 1 (PARP-1) activity, whose inhibition reduces RND1 transcription. In addition, overexpression of RND1 increases PARP-1, suggesting a cross-talk between PARP-1 and RND1. Finally, RND1 protects cells against camptothecin-induced apoptosis, and hence favors cellular resistance to camptothecin. Together, these findings highlight RND1 as an atypical RHO GTPase early induced by TOP1cc, and show that the TOP1cc-PARP-1-RND1 pathway protects cells against apoptosis induced by camptothecin.


Assuntos
Camptotecina/farmacologia , DNA Topoisomerases Tipo I/genética , DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Poli(ADP-Ribose) Polimerase-1/genética , Transcrição Gênica/genética , Proteínas rho de Ligação ao GTP/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Células HCT116 , Humanos , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Camundongos , Células NIH 3T3 , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores da Topoisomerase I/farmacologia
7.
Oncotarget ; 8(49): 86947-86968, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156849

RESUMO

Glioblastomas are malignant brain tumors with dismal prognosis despite standard treatment with surgery and radio/chemotherapy. These tumors are defined by an important cellular heterogeneity and notably contain a particular subpopulation of Glioblastoma-initiating cells, which recapitulate the heterogeneity of the original Glioblastoma. In order to classify these heterogeneous tumors, genomic profiling has also been undertaken to classify these heterogeneous tumors into several subtypes. Current research focuses on developing therapies, which could take into account this cellular and genomic heterogeneity. Among these targets, integrins are the subject of numerous studies since these extracellular matrix transmembrane receptors notably controls tumor invasion and progression. Moreover, some of these integrins are considered as membrane markers for the Glioblastoma-initiating cells subpopulation. We reviewed here integrin expression according to glioblastoma molecular subtypes and cell heterogeneity. We discussed their roles in glioblastoma invasion, angiogenesis, therapeutic resistance, stemness and microenvironment modulations, and provide an overview of clinical trials investigating integrins in glioblastomas. This review highlights that specific integrins could be identified as selective glioblastoma cells markers and that their targeting represents new diagnostic and/or therapeutic strategies.

8.
Mol Cell Biol ; 34(16): 3144-55, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24912678

RESUMO

Unlike other Rho GTPases, RhoB is rapidly induced by DNA damage, and its expression level decreases during cancer progression. Because inefficient repair of DNA double-strand breaks (DSBs) can lead to cancer, we investigated whether camptothecin, an anticancer drug that produces DSBs, induces RhoB expression and examined its role in the camptothecin-induced DNA damage response. We show that in camptothecin-treated cells, DSBs induce RhoB expression by a mechanism that depends notably on Chk2 and its substrate HuR, which binds to RhoB mRNA and protects it against degradation. RhoB-deficient cells fail to dephosphorylate γH2AX following camptothecin removal and show reduced efficiency of DSB repair by homologous recombination. These cells also show decreased activity of protein phosphatase 2A (PP2A), a phosphatase for γH2AX and other DNA damage and repair proteins. Thus, we propose that DSBs activate a Chk2-HuR-RhoB pathway that promotes PP2A-mediated dephosphorylation of γH2AX and DSB repair. Finally, we show that RhoB-deficient cells accumulate endogenous γH2AX and chromosomal abnormalities, suggesting that RhoB loss increases DSB-mediated genomic instability and tumor progression.


Assuntos
Quebras de DNA de Cadeia Dupla , Histonas/metabolismo , Proteína Fosfatase 2/metabolismo , Proteína rhoB de Ligação ao GTP/genética , Animais , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2/metabolismo , Aberrações Cromossômicas , Reparo do DNA/genética , Proteínas ELAV/metabolismo , Instabilidade Genômica/genética , Células HCT116 , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Ligação Proteica/genética , Proteína Fosfatase 2/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/metabolismo , Inibidores da Topoisomerase I/farmacologia , Proteína rhoB de Ligação ao GTP/biossíntese
9.
J Invest Dermatol ; 134(1): 203-212, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23792460

RESUMO

The role of UVB-induced apoptosis in the formation of squamous cell carcinoma (SCC) is recognized. We previously identified the small RhoB (Ras homolog gene family, member B) GTPase, an early response gene to cellular stress, as a critical protein controlling apoptosis of human keratinocytes after UVB exposure. Here we generated SKH1 (hairless immunocompetent mouse) mice invalidated for RhoB to evaluate its role in UVB-induced skin carcinogenesis in vivo. We show that rhob-/- mice have a lower risk of developing UVB-induced keratotic tumors and actinic keratosis that is associated with a higher sensitivity of UVB-exposed keratinocytes to apoptosis. We extend this observation to primary cultures of normal human keratinocytes in which RhoB was downregulated with small interfering RNA (siRNA) and further show that the hypersensitivity to apoptosis depends on B-cell lymphoma 2 (Bcl-2) downregulation. In rhob-/- mice, the UVB-induced tumors were preferentially undifferentiated and highly proliferative. Finally, we show in humans an almost constant loss of RhoB expression in undifferentiated SCCs. These undifferentiated and RhoB-deficient tumors have elevated phosphorylated histone H2AX (γH2AX) and 53BP1, two markers of DNA double-strand breaks. Together, our results indicate that UVB-induced RhoB expression participates in in vivo SCC initiation by increasing keratinocyte survival. Conversely, RhoB may limit tumor aggressiveness as loss of RhoB expression in tumor cells is associated with tumor progression.


Assuntos
Apoptose/fisiologia , Carcinoma de Células Escamosas/patologia , Queratinócitos/patologia , Neoplasias Cutâneas/patologia , Proteína rhoB de Ligação ao GTP/fisiologia , Animais , Apoptose/genética , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Queratinócitos/efeitos da radiação , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Pelados , Camundongos Knockout , RNA Interferente Pequeno/genética , Raios Ultravioleta/efeitos adversos , Proteína rhoB de Ligação ao GTP/genética
10.
Eur J Cancer ; 49(13): 2884-91, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23747271

RESUMO

We have previously shown that integrin-linked kinase (ILK) regulates U87 glioblastoma cell radioresistance by modulating the main radiation-induced cell death mechanism in solid tumours, the mitotic cell death. To decipher the biological pathways involved in these mechanisms, we constructed a U87 glioblastoma cell model expressing an inducible shRNA directed against ILK (U87shILK). We then demonstrated that silencing ILK enhanced radiation-induced centrosome overduplication, leading to radiation-induced mitotic cell death. In this model, ionising radiations induce hypoxia-inducible factor 1 alpha (HIF-1α) stabilisation which is inhibited by silencing ILK. Moreover, silencing HIF-1α in U87 cells reduced the surviving fraction after 2 Gy irradiation by increasing cell sensitivity to radiation-induced mitotic cell death and centrosome amplification. Because it is known that HIF-1α controls survivin expression, we then looked at the ILK silencing effect on survivin expression. We show that survivin expression is decreased in U87shILK cells. Furthermore, treating U87 cells with the specific survivin suppressor YM155 significantly increased the percentage of giant multinucleated cells, centrosomal overduplication and thus U87 cell radiosensitivity. In consequence, we decipher here a new pathway of glioma radioresistance via the regulation of radiation-induced centrosome duplication and therefore mitotic cell death by ILK, HIF-1α and survivin. This work identifies new targets in glioblastoma with the intention of radiosensitising these highly radioresistant tumours.


Assuntos
Glioblastoma/enzimologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Mitose/efeitos da radiação , Proteínas Serina-Treonina Quinases/metabolismo , Tolerância a Radiação , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Centrossomo/enzimologia , Centrossomo/patologia , Centrossomo/efeitos da radiação , Relação Dose-Resposta à Radiação , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos da radiação , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Transdução de Sinais/efeitos da radiação , Survivina , Fatores de Tempo , Transfecção
11.
J Cell Sci ; 125(Pt 18): 4264-77, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22718344

RESUMO

α6ß4 integrin is the main component of hemidesmosomes (HD) that stably anchor the epithelium to the underlying basement membrane. Epithelial cell migration requires HD remodelling, which can be promoted by epidermal growth factor (EGF). We previously showed that extracellular nucleotides inhibit growth factor-induced keratinocyte migration. Here, we investigate the effect of extracellular nucleotides on α6ß4 integrin localisation in HD during EGF-induced cell migration. Using a combination of pharmacological inhibition and gene silencing approaches, we found that UTP activates the P2Y2 purinergic receptor and Gαq protein to inhibit EGF/ERK1/2-induced cell migration in keratinocytes. Using a keratinocyte cell line expressing an inducible form of the Raf kinase, we show that UTP inhibits the EGF-induced ERK1/2 pathway activation downstream of Raf. Moreover, we established that ERK1/2 activation by EGF leads to the mobilisation of α6ß4 integrin from HD. Importantly, activation of P2Y2R and Gαq by UTP promotes HD formation and protects these structures from EGF-triggered dissolution as revealed by confocal analysis of the distribution of α6ß4 integrin, plectin, BPAG1, BPAG2 and CD151 in keratinocytes. Finally, we demonstrated that the activation of p90RSK, downstream of ERK1/2, is sufficient to promote EGF-mediated HD dismantling and that UTP does not stabilise HD in cells expressing an activated form of p90RSK. Our data underline an unexpected role of P2Y2R and Gαq in the inhibition of the ERK1/2 signalling pathway and in the modulation of hemidesmosome dynamics and keratinocyte migration.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Hemidesmossomos/metabolismo , Queratinócitos/citologia , Queratinócitos/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores Purinérgicos P2Y2/metabolismo , Movimento Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hemidesmossomos/efeitos dos fármacos , Humanos , Integrina beta4/metabolismo , Queratinócitos/efeitos dos fármacos , Modelos Biológicos , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Uridina Trifosfato/farmacologia , Quinases raf/metabolismo
12.
Mol Biol Cell ; 21(6): 946-55, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089844

RESUMO

Insulin-like growth factor-I (IGF-I) activation of phosphoinositol 3-kinase (PI3K) is an essential pathway for keratinocyte migration that is required for epidermis wound healing. We have previously reported that activation of Galpha((q/11))-coupled-P2Y(2) purinergic receptors by extracellular nucleotides delays keratinocyte wound closure. Here, we report that activation of P2Y(2) receptors by extracellular UTP inhibits the IGF-I-induced p110alpha-PI3K activation. Using siRNA and pharmacological inhibitors, we demonstrate that the UTP antagonistic effects on PI3K pathway are mediated by Galpha((q/11))-and not G((i/o))-independently of phospholipase Cbeta. Purinergic signaling does not affect the formation of the IGF-I receptor/insulin receptor substrate-I/p85 complex, but blocks the activity of a membrane-targeted active p110alpha mutant, indicating that UTP acts downstream of PI3K membrane recruitment. UTP was also found to efficiently attenuate, within few minutes, the IGF-I-induced PI3K-controlled translocation of the actin-nucleating protein cortactin to the plasma membrane. This supports the UTP ability to alter later migratory events. Indeed, UTP inhibits keratinocyte spreading and migration promoted by either IGF-I or a membrane-targeted active p110alpha mutant, in a Galpha(q/11)-dependent manner both. These findings provide new insight into the signaling cross-talk between receptor tyrosine kinase and Galpha((q/11))-coupled receptors, which mediate opposite effects on p110alpha-PI3K activity and keratinocyte migration.


Assuntos
Movimento Celular/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Queratinócitos/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Cortactina/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Humanos , Queratinócitos/citologia , Peptídeos Cíclicos/metabolismo , Fosfolipase C beta/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pseudópodes/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y2 , Uridina Trifosfato/metabolismo
13.
Cancer Res ; 69(8): 3308-16, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19351861

RESUMO

The presence of hypoxic areas in glioblastoma is an important determinant in tumor response to therapy and, in particular, to radiotherapy. Here we have explored the involvement of integrins, up to now known as regulators of angiogenesis and invasion, in the regulation of tumor hypoxia driven from the tumor cell. We first show that hypoxia induces the recruitment of alpha(v)beta(3) and alpha(v)beta(5) integrins to the cellular membrane of U87 and SF763 glioblastoma cells, thereby activating the focal adhesion kinase (FAK). We then show that inhibiting alpha(v)beta(3) or alpha(v)beta(5) integrins in hypoxic cells with a specific inhibitor or with siRNA decreases the hypoxia-inducible factor 1alpha (HIF-1alpha) intracellular level. This integrin-dependent regulation of HIF-1alpha is mediated through the regulation of FAK, which in turn activates the small GTPase RhoB, leading to the inhibition of GSK3-beta. Furthermore, silencing this pathway in glioma cells of established xenografts dramatically reduces glioma hypoxia, associated with a significant decrease in vessel density. Our present results unravel a new mechanism of hypoxia regulation by establishing the existence of an alpha(v)beta(3)/alpha(v)beta(5) integrin-dependent loop of hypoxia autoregulation in glioma. Targeting this hypoxia loop may be crucial to optimizing radiotherapy efficiency.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Glioblastoma/metabolismo , Integrina alfaVbeta3/metabolismo , Oxigênio/metabolismo , Receptores de Vitronectina/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Animais , Adesão Celular/fisiologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Glioblastoma/enzimologia , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrina alfaVbeta3/antagonistas & inibidores , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Vitronectina/antagonistas & inibidores , Transdução de Sinais , Transfecção , Transplante Heterólogo
14.
Int J Cancer ; 123(2): 357-364, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18464290

RESUMO

Integrins are extracellular matrix receptors involved in tumour invasion and angiogenesis. Although there is evidence that inhibiting integrins might enhance the efficiency of radiotherapy, little is known about the exact mechanisms involved in the integrin-dependent modulation of tumor radiosensitivity. The purpose of this study was to investigate the role of alphavbeta3 and alphavbeta5 integrins in glioblastoma cell radioresistance and overall to decipher the downstream biological pathways. We first demonstrated that silencing alphavbeta3 and alphavbeta5 integrins with specific siRNAs significantly reduced the survival after irradiation of 2 glioblastoma cell lines: U87 and SF763. We then showed that integrin activity and integrin signalling pathways controlled the glioma cell radiosensitivity. This regulation of glioma cell response to ionising radiation was mediated through the integrin-linked kinase, ILK, and the small GTPase, RhoB, by two mechanisms. The first one, independent of ILK, consists in the regulation of the intracellular level of RhoB by alphavbeta3 or alphavbeta5 integrin. The second pathway involved in cell radiosensitivity consists in RhoB activation by ionising radiation through ILK. Furthermore, we demonstrated that the alphavbeta3/alphavbeta5 integrins/ILK/RhoB pathway controlled the glioma cells radiosensitivity by regulating radiation-induced mitotic cell death. This work identifies a new biological pathway controlling glioblastoma cells radioresistance, activated from the membrane through alphavbeta3 and/or alphavbeta5 integrins via ILK and RhoB. Our results are clues that downstream effectors of alphavbeta3 and alphavbeta5 integrins as ILK and RhoB might also be promising candidate targets for improving the efficiency of radiotherapy and thus the clinical outcome of patients with glioblastoma.


Assuntos
Glioma/metabolismo , Glioma/radioterapia , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Radiação Ionizante , Receptores de Vitronectina/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral/efeitos da radiação , Citometria de Fluxo , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , RNA Interferente Pequeno/metabolismo , Tolerância a Radiação , Transfecção
15.
Cancer Res ; 66(1): 482-9, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397264

RESUMO

Hypoxia is a crucial factor in tumor aggressiveness and resistance to treatment, particularly in glioma. Our previous results have shown that inhibiting the small GTPase RhoB increased oxygenation of U87 human glioblastoma xenografts, in part, by regulating angiogenesis. We investigated here whether RhoB might also control a signaling pathway that would permit glioma cells to adapt to hypoxia. We first showed that silencing RhoB with siRNA induced degradation and inhibition of the transcriptional activity of the hypoxia-inducible factor by the proteasome in U87 hypoxic cells. This RhoB-dependent degradation of hypoxia-inducible factor-1alpha in hypoxic conditions was mediated by the Akt/glycogen synthase kinase-3beta pathway. While investigating how hypoxia could activate this signaling pathway, using the GST-Rhotekin RBD pulldown assay, we showed the early activation of RhoB by reactive oxygen species under hypoxic conditions and, subsequently, its participation in the ensuing cellular adaptation to hypoxia. Overall, therefore, our results have not only highlighted a new signaling pathway for hypoxia controlled by the small GTPase RhoB, but they also strongly implicate RhoB as a potentially important therapeutic target for decreasing tumor hypoxia.


Assuntos
Glioblastoma/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Ativação Enzimática , Glioblastoma/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteína rhoB de Ligação ao GTP/antagonistas & inibidores
16.
Cell Cycle ; 4(3): 438-41, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15738653

RESUMO

The Ku heterodimer (Ku70/Ku80) plays a central role in DNA double strand break recognition and repair. It has been shown, more than ten years ago, that Ku is also expressed at the cell surface of different cells types along with its intracellular pool within the nucleus and the cytoplasm but involvement of Ku in cell-cell and cell-extracellular matrix adhesion has been only recently demonstrated. In addition, we have shown that Ku may have a second and unexpected activity in cell/microenvironment interaction. Indeed, Ku appears to be involved in extracellular proteolytic processes through its specific interaction, on the cell surface, with the matrix metalloprotease 9. Taken together, these results suggest that Ku function at the cell surface is likely to be important in tumour invasion. Various fundamental questions arise from these observations. How Ku is expressed on the cell surface, why a protein with completely unrelated functions also serve as an integrin-like molecule once expressed at the cell surface and is this functional moonlighting of Ku related to cell transformation remain open issues that will be discussed here.


Assuntos
Antígenos Nucleares/fisiologia , Dano ao DNA , Proteínas de Ligação a DNA/fisiologia , Animais , Adesão Celular , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Reparo do DNA , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Autoantígeno Ku , Metaloproteinase 9 da Matriz/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Ligação Proteica
17.
EMBO J ; 23(19): 3758-68, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15385961

RESUMO

The Ku heterodimer (Ku70/Ku80) plays a central role in DNA double-strand breaks repair. Ku is also expressed on the cell surface of different types of cells where its function remains poorly understood. From a yeast two-hybrid screen, we have identified a specific interaction between the core region of Ku80 and the hemopexin domain of metalloproteinase 9 (MMP-9), a key enzyme involved in the degradation of extracellular matrix (ECM) components. Ku associates with MMP-9 on the surface of leukemic cells as demonstrated by co-immunoprecipitation experiments in membrane extracts and double-label immunofluorescence studies. In normal and tumoral migratory cells, Ku80 and MMP-9 colocalize at the periphery of leading edge of cells and cellular invasion of collagen IV matrices was blocked by antibodies directed against Ku70 or Ku80 subunits as well as by Ku80-specific antisense oligonucleotides. Our results indicate that Ku and MMP-9 interact at the cell membrane of highly invasive hematopoietic cells of normal and tumoral origin and document the unexpected importance of the membrane-associated form of Ku in the regulation of ECM remodelling.


Assuntos
Antígenos Nucleares/metabolismo , Membrana Celular/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Antígenos Nucleares/genética , Movimento Celular , Colágeno Tipo IV/metabolismo , Proteínas de Ligação a DNA/genética , Imunofluorescência , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunoprecipitação , Autoantígeno Ku , Leucemia/metabolismo , Leucemia/patologia , Invasividade Neoplásica , Oligonucleotídeos Antissenso/farmacologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
18.
J Mol Biol ; 337(3): 503-11, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-15019772

RESUMO

The Ku heterodimer (Ku70/Ku80) plays a central role in DNA double-strand breaks recognition and repair. However, Ku is expressed also on the surface of different types of cells along with its intracellular pool within the nucleus and the cytoplasm. Participation of membrane-associated Ku in cell-cell interaction has been reported recently. Here, we describe a novel function of cell-surface Ku as an adhesion receptor for fibronectin (Fn). The role of Ku in cell adhesion was investigated by comparing the Ku80 deficient Chinese hamster ovary (CHO) cell line, xrs-6, with clones transfected stably with either the hamster or human Ku80 cDNA. Ku expression in transfectant cells resulted in a significant increased adhesion on Fn and type IV collagen as compared to control cells. The observed increase in cell adhesion relied on Ku cell-surface expression, since antibodies directed against Ku70 or Ku80 subunit inhibited adhesion on Fn of Ku80, but not control vector, transfected xrs-6 cells. In addition, both Ku70 and Ku80 present a structural relationship with integrin I (or A) domains and the A1 and A3 domains of von Willebrand factor, domains known to be involved in Fn binding. Both Ku70 and Ku80 exhibit a complete set of residues compatible in their position and chemical nature with the formation of a metal ion-dependent adhesion (MIDAS) site implicated in ligand binding and integrin activation. Taken together, these functional and structural approaches support a new role for Ku as an adhesion receptor for Fn.


Assuntos
Antígenos Nucleares/fisiologia , Adesão Celular , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Fibronectinas/metabolismo , Proteínas de Membrana/fisiologia , Sequência de Aminoácidos , Animais , Antígenos Nucleares/química , Antígenos Nucleares/genética , Células CHO , Cricetinae , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Humanos , Integrinas/química , Autoantígeno Ku , Proteínas de Membrana/química , Mesocricetus , Modelos Moleculares , Receptores de Fibronectina , Alinhamento de Sequência , Homologia Estrutural de Proteína , Transfecção , Fator de von Willebrand/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...