Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 22(1): 843, 2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35918659

RESUMO

BACKGROUND: Glioblastoma (GB) is the most common and most aggressive malignant brain tumor. In understanding its resistance to conventional treatments, iron metabolism and related pathways may represent a novel avenue. As for many cancer cells, GB cell growth is dependent on iron, which is tightly involved in red-ox reactions related to radiotherapy effectiveness. From new observations indicating an impact of RX radiations on the expression of ceruloplasmin (CP), an important regulator of iron metabolism, the aim of the present work was to study the functional effects of constitutive expression of CP within GB lines in response to beam radiation depending on the oxygen status (21% O2 versus 3% O2). METHODS AND RESULTS: After analysis of radiation responses (Hoechst staining, LDH release, Caspase 3 activation) in U251-MG and U87-MG human GB cell lines, described as radiosensitive and radioresistant respectively, the expression of 9 iron partners (TFR1, DMT1, FTH1, FTL, MFRN1, MFRN2, FXN, FPN1, CP) were tested by RTqPCR and western blots at 3 and 8 days following 4 Gy irradiation. Among those, only CP was significantly downregulated, both at transcript and protein levels in the two lines, with however, a weaker effect in the U87-MG, observable at 3% O2. To investigate specific role of CP in GB radioresistance, U251-MG and U87-MG cells were modified genetically to obtain CP depleted and overexpressing cells, respectively. Manipulation of CP expression in GB lines demonstrated impact both on cell survival and on activation of DNA repair/damage machinery (γH2AX); specifically high levels of CP led to increased production of reactive oxygen species, as shown by elevated levels of superoxide anion, SOD1 synthesis and cellular Fe2 + . CONCLUSIONS: Taken together, these in vitro results indicate for the first time that CP plays a positive role in the efficiency of radiotherapy on GB cells.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Ceruloplasmina/farmacologia , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Ferro/farmacologia , Oxigênio/metabolismo , Tolerância a Radiação/genética
2.
BMC Cancer ; 16: 51, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26832741

RESUMO

BACKGROUND: Overcoming resistance to treatment is an essential issue in many cancers including glioblastoma (GBM), the deadliest primary tumor of the central nervous system. As dependence on iron is a key feature of tumor cells, using chelators to reduce iron represents an opportunity to improve conventional GBM therapies. The aim of the present study was, therefore, to investigate the cytostatic and cytotoxic impact of the new iron chelator deferasirox (DFX) on human GBM cells in well-defined clinical situations represented by radiation therapy and mild-hypoxia. RESULTS: Under experimental normoxic condition (21% O2), deferasirox (DFX) used at 10 µM for 3 days reduced proliferation, led cell cycle arrest in S and G2-M phases and induced cytotoxicity and apoptosis in U251 and U87 GBM cells. The abolition of the antineoplastic DFX effects when cells were co-treated with ferric ammonium sulfate supports the hypothesis that its effects result from its ability to chelate iron. As radiotherapy is the main treatment for GBM, the combination of DFX and X-ray beam irradiation was also investigated. Irradiation at a dose of 16 Gy repressed proliferation, cytotoxicity and apoptosis, but only in U251 cells, while no synergy with DFX was observed in either cell line. Importantly, when the same experiment was conducted in mild-hypoxic conditions (3% O2), the antiproliferative and cytotoxic effects of DFX were abolished, and its ability to deplete iron was also impaired. CONCLUSIONS: Taken together, these in vitro results could raise the question of the benefit of using iron chelators in their native forms under the hypoxic conditions often encountered in solid tumors such as GBM. Developing new chemistry or a new drug delivery system that would keep DFX active in hypoxic cells may be the next step toward their application.


Assuntos
Benzoatos/administração & dosagem , Hipóxia Celular , Glioblastoma/metabolismo , Quelantes de Ferro/administração & dosagem , Triazóis/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada , Deferasirox , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Oxigênio/metabolismo
3.
EBioMedicine ; 105: 105202, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38905749

RESUMO

BACKGROUND: Glioblastoma (GB), the most aggressive brain cancer, remains a critical clinical challenge due to its resistance to conventional treatments. Here, we introduce a locoregional targeted-α-therapy (TAT) with the rat monoclonal antibody 9E7.4 targeting murine syndecan-1 (SDC1) coupled to the α-emitter radionuclide astatine-211 (211At-9E7.4). METHODS: We orthotopically transplanted 50,000 GL261 cells of murine GB into the right striatum of syngeneic female C57BL/6JRj mice using stereotaxis. After MRI validation of tumour presence at day 11, TAT was injected at the same coordinates. Biodistribution, efficacy, toxicity, local and systemic responses were assessed following application of this protocol. The 9E7.4 monoclonal antibody was labelled with iodine-125 (125I) for biodistribution and with astatine-211 (211At) for the other experiments. FINDINGS: The 211At-9E7.4 TAT demonstrated robust efficacy in reducing orthotopic tumours and achieved improved survival rates in the C57BL/6JRj model, reaching up to 70% with a minimal activity of 100 kBq. Targeting SDC1 ensured the cerebral retention of 211At over an optimal time window, enabling low-activity administration with a minimal toxicity profile. Moreover, TAT substantially reduced the occurrence of secondary tumours and provided resistance to new tumour development after contralateral rechallenge, mediated through the activation of central and effector memory T cells. INTERPRETATION: The locoregional 211At-9E7.4 TAT stands as one of the most efficient TAT across all preclinical GB models. This study validates SDC1 as a pertinent therapeutic target for GB and underscores 211At-9E7.4 TAT as a promising advancement to improve the treatment and quality of life for patients with GB. FUNDING: This work was funded by the French National Agency for Research (ANR) "France 2030 Investment Plan" Labex Iron [ANR-11-LABX-18-01], The SIRIC ILIAD [INCa-DGOS-INSERM-18011], the French program "Infrastructure d'Avenir en Biologie-Santé" (France Life Imaging) [ANR-11-INBS-0006], the PIA3 of the ANR, integrated to the "France 2030 Investment Plan" [ANR-21-RHUS-0012], and support from Inviscan SAS (Strasbourg, France). It was also related to: the ANR under the frame of EuroNanoMed III (project GLIOSILK) [ANR-19-ENM3-0003-01]; the "Région Pays-de-la-Loire" under the frame of the Target'In project; the "Ligue Nationale contre le Cancer" and the "Comité Départemental de Maine-et-Loire de la Ligue contre le Cancer" (CD49) under the frame of the FusTarG project and the "Tumour targeting, imaging and radio-therapies network" of the "Cancéropôle Grand-Ouest" (France). This work was also funded by the Institut National de la Santé et de la Recherche Médicale (INSERM), the University of Nantes, and the University of Angers.

4.
Acta Biomater ; 173: 261-282, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37866725

RESUMO

In view of inevitable recurrences despite resection, glioblastoma (GB) is still an unmet clinical need. Dealing with the stromal-cell derived factor 1-alpha (SDF-1α)/CXCR4 axis as a hallmark of infiltrative GB tumors and with the resection cavity situation, the present study described the effects and relevance of a new engineered micro-nanostructured SF-HA-Hep aerogel sponges, made of silk fibroin (SF), hyaluronic acid (HA) and heparin (Hep) and loaded with SDF-1α, to interfere with the GB ecosystem and residual GB cells, attracting and confining them in a controlled area before elimination. 70 µm-pore sponges were designed as an implantable scaffold to trap GB cells. They presented shape memory and fit brain cavities. Histological results after implantation in brain immunocompetent Fischer rats revealed that SF-HA-Hep sponges are well tolerated for more than 3 months while moderately and reversibly colonized by immuno-inflammatory cells. The use of human U87MG GB cells overexpressing the CXCR4 receptor (U87MG-CXCR4+) and responding to SDF-1α allowed demonstrating directional GB cell attraction and colonization of the device in vitro and in vivo in orthotopic resection cavities in Nude rats. Not modifying global survival, aerogel sponge implantation strongly shaped U87MG-CXCR4+ tumors in cavities in contrast to random infiltrative growth in controls. Overall, those results support the interest of SF-HA-Hep sponges as modifiers of the GB ecosystem dynamics acting as "cell meeting rooms" and biocompatible niches whose properties deserve to be considered toward the development of new clinical procedures. STATEMENT OF SIGNIFICANCE: Brain tumor glioblastoma (GB) is one of the worst unmet clinical needs. To prevent the relapse in the resection cavity situation, new implantable biopolymer aerogel sponges loaded with a chemoattractant molecule were designed and preclinically tested as a prototype targeting the interaction between the initial tumor location and its attraction by the peritumoral environment. While not modifying global survival, biocompatible SDF1-loaded hyaluronic acid and silk fibroin sponges induce directional GB cell attraction and colonization in vitro and in rats in vivo. Interestingly, they strongly shaped GB tumors in contrast to random infiltrative growth in controls. These results provide original findings on application of exogenous engineered niches that shape tumors and serve as cell meeting rooms for further clinical developments.


Assuntos
Neoplasias Encefálicas , Fibroínas , Glioblastoma , Ratos , Humanos , Animais , Quimiocina CXCL12/farmacologia , Fibroínas/farmacologia , Ácido Hialurônico/farmacologia , Ecossistema , Recidiva Local de Neoplasia , Neoplasias Encefálicas/cirurgia , Receptores CXCR4
5.
Polymers (Basel) ; 13(4)2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-33578913

RESUMO

Polysaccharides have received a lot of attention in biomedical research for their high potential as scaffolds owing to their unique biological properties. Fibrillar scaffolds made of chitosan demonstrated high promise in tissue engineering, especially for skin. As far as bone regeneration is concerned, curdlan (1,3-ß-glucan) is particularly interesting as it enhances bone growth by helping mesenchymal stem cell adhesion, by favoring their differentiation into osteoblasts and by limiting the osteoclastic activity. Therefore, we aim to combine both chitosan and curdlan polysaccharides in a new scaffold for bone regeneration. For that purpose, curdlan was electrospun as a blend with chitosan into a fibrillar scaffold. We show that this novel scaffold is biodegradable (8% at two weeks), exhibits a good swelling behavior (350%) and is non-cytotoxic in vitro. In addition, the benefit of incorporating curdlan in the scaffold was demonstrated in a scratch assay that evidences the ability of curdlan to express its immunomodulatory properties by enhancing cell migration. Thus, these innovative electrospun curdlan-chitosan scaffolds show great potential for bone tissue engineering.

6.
Front Bioeng Biotechnol ; 8: 602998, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33718332

RESUMO

Inhibition of the PI3K/Akt/mTOR signaling pathway represents a potential issue for the treatment of cancer, including glioblastoma. As such, rapamycin that inhibits the mechanistic target of rapamycin (mTOR), the downstream effector of this signaling pathway, is of great interest. However, clinical development of rapamycin has floundered due to the lack of a suitable formulation of delivery systems. In the present study, a novel method for the formulation of safe rapamycin nanocarriers is investigated. A phase inversion process was adapted to prepare lipid nanocapsules (LNCs) loaded with the lipophilic and temperature sensitive rapamycin. Rapamycin-loaded LNCs (LNC-rapa) are ~110 nm in diameter with a low polydispersity index (<0.05) and the zeta potential of about -5 mV. The encapsulation efficiency, determined by spectrophotometry conjugated with filtration/exclusion, was found to be about 69%, which represents 0.6 wt% of loading capacity. Western blot analysis showed that LNC-rapa do not act synergistically with X-ray beam radiation in U87MG glioblastoma model in vitro. Nevertheless, it demonstrated the selective inhibition of the phosphorylation of mTORC1 signaling pathway on Ser2448 at a concentration of 1 µM rapamycin in serum-free medium. Interestingly, cells cultivated in normoxia (21% O2) seem to be more sensitive to mTOR inhibition by rapamycin than those cultivated in hypoxia (0.4% O2). Finally, we also established that mTOR phosphorylation inhibition by LNC-rapa induced a negative feedback through the activation of Akt phosphorylation. This phenomenon was more noticeable after stabilization of HIF-1α in hypoxia.

7.
Mol Biol Cell ; 16(4): 1850-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15659654

RESUMO

Besides its function as a cell cycle regulator, cyclin D1 interacts with transcription factors to regulate gene activation. In this study, we show that cyclin D1 is recruited to the p21waf1 promoter by a STAT3-NcoA complex. The association of cyclin D1 with DNA prevented the recruitment of the CBP histone acetylase and RNA polymerase II, leading to an inhibition of the p21waf1 gene. Confirming the transcriptional function of the protein, the expression of the p21waf1 gene was enhanced in cyclin D1-/- fibroblasts or upon siRNA-mediated down-regulation of the cyclin. Moreover, the STAT3-mediated activation of p21waf1 was also inhibited in breast cancer cells containing elevated levels of cyclin D1. Altogether, these results suggest that the transcriptional activities of cyclin D1 might play an important role in the regulation of cell-cycle regulatory genes and that these functions are probably involved in cell transformation.


Assuntos
Ciclina D1/metabolismo , DNA/metabolismo , Regulação da Expressão Gênica/genética , Transcrição Gênica/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteína de Ligação a CREB , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina D1/deficiência , Ciclina D1/genética , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Polimerase II/metabolismo , Fator de Transcrição STAT3 , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional
8.
Biomater Sci ; 6(9): 2386-2409, 2018 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-30023990

RESUMO

Glioblastoma is the most frequent and aggressive primary malignant tumor of the central nervous system with a gloomy prognosis. Platinum derivatives and one among them, cisplatin, exhibited promising results when locally administered into the brain of glioblastoma bearing rats. Nanovectorization of anticancer agents through polymeric nanoparticles may even promote drug accumulation within cells, thus concentrating the drug efficiently at its target. Anchorage of gadolinium complexes on the corona of such smart drug delivery systems could further allow magnetic resonance imaging (MRI) monitoring of the nanoplatform biodistribution in the damaged parenchyma and its therapeutic benefit. For this purpose, a biocompatible amphiphilic triblock copolymer, made of degradable polyester and polycarbonate and bioeliminable polyethylene oxide (PEO), was synthesized by successive ring-opening polymerizations. After micellization in water, gadolinium complexes were grafted onto the PEO micelle corona and the carboxylate functions, located at the surface of the micelle's core, were able to cross-link with Pt(ii) complexes. A macromolecular prodrug was therefore recovered in which more than one third of the carboxylate functions were linked to a platinum atom. By this strategy, stable cisplatin cross-linked nanoparticles were formulated with a mean size in the range of 100.63 ± 12.04 nm consistent with biological investigations. Relaxometry measurements both in water and in plasma at 7 T, 25 °C, confirmed the intrinsic potential of these hybrid nanoparticles as alternative MRI contrast agents with a substantial increase in the r2/r1 ratio by a factor of 3.3 and 2.7, respectively, compared to the conventional low molar mass Gd-DTPA. As a result, their infusion within the striatum of glioblastoma-bearing mice resulted in a hypersignal on T2-weighted MR images that persisted over time. Ultimately, the formulated prodrug exhibited up to 50-fold increased accumulation in human glioblastoma cell lines and up to 32-fold enhanced subsequent Pt-DNA adduct formation in comparison with free cisplatin, thus supporting the potential of this innovative bimodal tool for further applications.


Assuntos
Antineoplásicos , Cisplatino , Gadolínio DTPA , Nanopartículas , Pró-Fármacos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/química , Adutos de DNA/metabolismo , Liberação Controlada de Fármacos , Feminino , Gadolínio DTPA/administração & dosagem , Gadolínio DTPA/química , Glioblastoma/tratamento farmacológico , Camundongos Nus , Nanopartículas/administração & dosagem , Nanopartículas/química , Platina/metabolismo , Cimento de Policarboxilato/química , Poliésteres/administração & dosagem , Poliésteres/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química
9.
Theranostics ; 7(18): 4517-4536, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29158842

RESUMO

PURPOSE: Gold standard beam radiation for glioblastoma (GBM) treatment is challenged by resistance phenomena occurring in cellular populations well prepared to survive or to repair damage caused by radiation. Among signals that have been linked with radio-resistance, the SDF1/CXCR4 axis, associated with cancer stem-like cell, may be an opportune target. To avoid the problem of systemic toxicity and blood-brain barrier crossing, the relevance and efficacy of an original system of local brain internal radiation therapy combining a radiopharmaceutical with an immuno-nanoparticle was investigated. EXPERIMENT DESIGN: The nanocarrier combined lipophilic thiobenzoate complexes of rhenium-188 loaded in the core of a lipid nanocapsule (LNC188Re) with a function-blocking antibody, 12G5 directed at the CXCR4, on its surface. The efficiency of 12G5-LNC188Re was investigated in an orthotopic and xenogenic GBM model of CXCR4-positive U87MG cells implanted in the striatum of Scid mice. RESULTS: We demonstrated that 12G5-LNC188Re single infusion treatment by convection-enhanced delivery resulted in a major clinical improvement in median survival that was accompanied by locoregional effects on tumor development including hypovascularization and stimulation of the recruitment of bone marrow derived CD11b- or CD68-positive cells as confirmed by immunohistochemistry analysis. Interestingly, thorough analysis by spectral imaging in a chimeric U87MG GBM model containing CXCR4-positive/red fluorescent protein (RFP)-positive- and CXCR4-negative/RFP-negative-GBM cells revealed greater confinement of DiD-labeled 12G5-LNCs than control IgG2a-LNCs in RFP compartments. Main conclusion: These findings on locoregional impact and targeting of disseminated cancer cells in tumor margins suggest that intracerebral active targeting of nanocarriers loaded with radiopharmaceuticals may have considerable benefits in clinical applications.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Nanopartículas/administração & dosagem , Radioisótopos/administração & dosagem , Compostos Radiofarmacêuticos/administração & dosagem , Receptores CXCR4/administração & dosagem , Rênio/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos da radiação , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Lipídeos/administração & dosagem , Camundongos , Nanocápsulas/administração & dosagem , Células-Tronco Neoplásicas/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
10.
Oncogene ; 23(44): 7391-8, 2004 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-15286705

RESUMO

STAT transcription factors (signal transducers and activators of transcription) are cytoplasmic proteins that induce gene activation in response to cytokine receptor stimulation. Following tyrosine phosphorylation, STAT proteins translocate into the nucleus and activate specific target genes. We have previously reported that STAT3 activates the expression of the p21waf1 gene through its association with the NcoA/SRC1a and CBP coactivators. In this study, we explore the role of BRG1, a component of the SWI/SNF chromatin-remodeling complex, and the role of cdk9, a component of the elongation factor P-TEFb, in the STAT3-mediated expression of p21waf1. We found using pull-down experiments and co-immunoprecipitation assays that both proteins associate with STAT3. Chromatin immunoprecipitation (ChIP) experiments indicate that STAT3 DNA binding results in histone H3 acetylation and BRG1 recruitment. Using Southern blot analysis, we found that the loading of BRG1 is followed by an increased accessibility of the proximal p21waf1 promoter and by the association of RNA polymerase II. As a next step, STAT3 then recruits the cdk9 kinase to phosphorylate the carboxy-terminal domain of the RNA polymerase at serine 2. Accordingly, the elongating form of the polymerase can be detected by ChIP experiments on the coding region of the gene, probably initiating mRNA synthesis. Therefore, STAT3 not only promotes the initiation of transcription but also regulates chromatin remodeling and transcription elongation through its interaction with BRG1 and cdk9.


Assuntos
Quinase 9 Dependente de Ciclina/metabolismo , Ciclinas/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21 , DNA Helicases , Éxons/genética , Regulação da Expressão Gênica , Humanos , Interleucina-6/farmacologia , Regiões Promotoras Genéticas/genética , Fator de Transcrição STAT3 , Transcrição Gênica/genética , Ativação Transcricional
11.
J Biol Chem ; 278(5): 2990-6, 2003 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-12438313

RESUMO

Activated forms of STAT3 transcription factors are often found in various cancers and tumor cell lines, indicating that this signaling pathway is involved in tumorogenesis. At the molecular level, STAT3 proteins function as transcriptional activators and up-regulate several growth-promoting genes such as myc, pim-1, or cyclin D1. However, these transcription factors have also proapoptotic functions and can activate the expression of the cell-cycle inhibitor p21(waf1), suggesting that STAT3 can also block cell-cycle progression and prevent abnormal cell proliferation. To reconcile these observations, one would predict that the STAT3-mediated activation of p21(waf1) is lost during cell transformation. In this study, we show that upon IL-6 stimulation of glioblastoma cells, STAT3 does not activate the expression of the p21(waf1) gene, whereas the expression of the myc gene remains unaltered. Chromatin immunoprecipitation experiments show that STAT3 and its cofactor NcoA/SRC1a are effectively recruited to the p21(waf1) promoter but that this is not followed by the association of the CREB-binding protein (CBP) histone acetylase and the type II RNA polymerase as normally seen on the myc promoter. Whereas the PI-3K/Akt pathway is constitutively activated in these cells, inactivation of this pathway restores the loading of CBP and the RNA polymerase and the expression of the p21(waf1) gene without having any effect on myc regulation. Moreover, this effect was recapitulated in HepG2 cells expressing an activated form of the Akt kinase. In these cells, the kinase blocked the STAT3-mediated expression of the p21(waf1) gene by inhibiting the recruitment of CREB-binding protein and the type II RNA polymerase, without having any effects on the loading of STAT3 and its cofactor NcoA/SRC1a. Together, these findings suggest that the phosphatidylinositol 3-kinase/Akt pathway inhibits the transcriptional activation of the p21(waf1) gene by STAT3 proteins without altering the regulation of the myc promoter.


Assuntos
Ciclinas/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Transativadores/fisiologia , Animais , Sequência de Bases , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Linhagem Celular , Transformação Celular Neoplásica , Cromatina/genética , Cromatina/fisiologia , Meios de Cultura Livres de Soro , Inibidor de Quinase Dependente de Ciclina p21 , Sondas de Oligonucleotídeos , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT3 , Fatores de Transcrição/metabolismo , Transfecção
12.
Blood ; 101(10): 4070-7, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12531804

RESUMO

DNA viruses have evolved a number of mechanisms to inhibit the major cellular tumor-suppressor pathways. Viral oncogenes can override growth suppressive signals and extend the virus proliferative capacity. The Kaposi sarcoma-associated human herpesvirus 8 (KSHV) encodes a protein, cyclin K, that is similar to cellular cyclin D1 but behaves atypically. Cyclin K resists the actions of the p16 INK4a and p27Kip1 inhibitors and extends the range of cdk6 substrates, thereby inducing cell-cycle progression toward S phase. In this study, we show that cyclin K overrides growth suppressive signals through signal transducer and activator of transcription 3 (STAT3) inactivation. Cyclin K was found to associate with the activation domain of STAT3 to inhibit its DNA-binding and transcriptional activities. Overexpression of cyclin K and inhibition of STAT3 prevents the growth suppressive effect imposed by the interleukin 6-type cytokine, oncostatin M. Altogether, these results suggest that KSHV is able to override growth suppressive effects through multiple mechanisms, and they further indicate that cyclin K plays an important role in the oncogenic activity of these viruses.


Assuntos
Ciclinas/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Inibidores do Crescimento/farmacologia , Herpesvirus Humano 8/fisiologia , Peptídeos/farmacologia , Sarcoma de Kaposi/virologia , Transativadores/antagonistas & inibidores , Proteínas Virais/farmacologia , Divisão Celular/efeitos dos fármacos , Humanos , Melanoma , Oncostatina M , Peptídeos/antagonistas & inibidores , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Fator de Transcrição STAT3 , Sarcoma de Kaposi/patologia , Células Tumorais Cultivadas
13.
J Biol Chem ; 277(10): 8004-11, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11773079

RESUMO

Signal transducer and activator of transcription 3 (STAT3) transcription factors are cytoplasmic proteins that induce gene activation in response to cytokine receptor stimulation. Following tyrosine phosphorylation, STAT3 proteins dimerize, translocate to the nucleus, and activate specific target genes. This transcriptional activation by STAT3 proteins has been shown to require the recruitment of coactivators such as CREB-binding protein (CBP)/p300. In the present study, we show that steroid receptor coactivator 1, NcoA/SRC1a, originally identified as a nuclear receptor coactivator, also functions as a coactivator of STAT3 proteins. In coimmunoprecipitations, NcoA/SRC1a was found to associate with STAT3 following IL-6 stimulation of HepG2 hepatoma cells. Pull-down experiments indicated that the N-terminal part of NcoA/SRC1a associates with the activation domain of STAT3. Overexpression of NcoA/SRC1a or its SRC1e isoform enhanced transcriptional activation by STAT3 proteins in transient transfection experiments. This ability of NcoA/SRC1a to enhance STAT3 activity is dependent upon the presence of the CBP-interacting domain, activation domain 1. Using chromatin immunoprecipitation assays, we found that STAT3, NcoA/SRC1a, and CBP/p300 are simultaneously recruited to the p21(waf1) promoter following interleukin-6 stimulation. Taken together, these data suggest that CBP/p300 and NcoA/SRC1a may function in a common pathway to regulate STAT3 transcriptional activity.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Transativadores/química , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Western Blotting , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ciclina D1/metabolismo , DNA Complementar/metabolismo , Dimerização , Glutationa Transferase/metabolismo , Histona Acetiltransferases , Histonas/metabolismo , Humanos , Immunoblotting , Interleucina-6/metabolismo , Luciferases/metabolismo , Dados de Sequência Molecular , Coativador 1 de Receptor Nuclear , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Fator de Transcrição STAT3 , Transcrição Gênica , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa