Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Hum Mol Genet ; 28(21): 3610-3624, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31511867

RESUMO

Mucopolysaccharidosis (MPS) type VII is a lysosomal storage disease caused by ß-glucuronidase deficiency, prompting glycosaminoglycan accumulation in enlarged vesicles, leading to peripheral and neuronal dysfunction. Here, we present a gene therapy strategy using lumbar puncture of AAVrh10 encoding human ß-glucuronidase (AAVrh10-GUSB) to adult MPS VII mice. This minimally invasive technique efficiently delivers the recombinant vector to the cerebrospinal fluid (CSF) with a single intrathecal injection. We show that AAVrh10 delivery to the CSF allows global, stable transduction of CNS structures. In addition, drainage of AAVrh10-GUSB from the CSF to the bloodstream resulted in the transduction of somatic organs such as liver, which provided a systemic ß-glucuronidase source sufficient to achieve serum enzyme activity comparable to wild type mice. ß-glucuronidase levels were enough to correct biochemical and histopathological hallmarks of the disease in the CNS and somatic organs at short and long term. Moreover, the progression of the bone pathology was also reduced. Importantly, the biochemical correction led to a significant improvement in the physical, cognitive and emotional characteristics of MPS VII mice, and doubling their life span. Our strategy may have implications for gene therapy in patients with lysosomal storage diseases.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/terapia , Animais , Comportamento Animal , Cognição , Dependovirus/metabolismo , Modelos Animais de Doenças , Emoções , Vetores Genéticos/metabolismo , Glucuronidase/administração & dosagem , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Mucopolissacaridose VII/mortalidade , Mucopolissacaridose VII/psicologia , Sobrevida
2.
Oncogenesis ; 6(7): e354, 2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-28671677

RESUMO

The proximity of organs at risk makes the treatment of head and neck squamous cell carcinoma (HNSCC) challenging by standard radiotherapy. The higher precision in tumor targeting of proton (P) therapy could promote it as the treatment of choice for HNSCC. Besides the physical advantage in dose deposition, few is known about the biological impact of P versus photons (X) in this setting. To investigate the comparative biological effects of P versus X radiation in HNSCC cells, we assessed the relative biological effectiveness (RBE), viability, proliferation and mRNA levels for genes involved in (lymph)angiogenesis, inflammation, proliferation and anti-tumor immunity. These parameters, particularly VEGF-C protein levels and regulations, were documented in freshly irradiated and/or long-term surviving cells receiving low/high-dose, single (SI)/multiple (MI) irradiations with P/X. The RBE was found to be 1.1 Key (lymph)angiogenesis and inflammation genes were downregulated (except for vegf-c) after P and upregulated after X irradiation in MI surviving cells, demonstrating a more favorable profile after P irradiation. Both irradiation types stimulated vegf-c promoter activity in a NF-κB-dependent transcriptional regulation manner, but at a lesser extent after P, as compared to X irradiation, which correlated with mRNA and protein levels. The cells surviving to MI by P or X generated tumors with higher volume, anarchic architecture and increased density of blood vessels. Increased lymphangiogenesis and a transcriptomic analysis in favor of a more aggressive phenotype were observed in tumors generated with X-irradiated cells. Increased detection of lymphatic vessels in relapsed tumors from patients receiving X radiotherapy was consistent with these findings. This study provides new data about the biological advantage of P, as compared to X irradiation. In addition to its physical advantage in dose deposition, P irradiation may help to improve treatment approaches for HNSCC.

3.
Oral Oncol ; 57: 46-53, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27208844

RESUMO

The "Hallmarks of Cancer" describe the ways by which cancer cells bypass homeostasis. Escape from replicative senescence is one of the earliest features of cancer cells. Maintenance of the telomeres through reactivation of telomerase was initially associated with replicative immortality in various cancers. The shelterin complex, a telomeric hexaprotein association, plays a key role in telomere maintenance and in the hallmarks of cancer. Some shelterin proteins are overexpressed in diverse cancers and can promote tumorigenesis in animal models. Shelterin can also have an impact on tumor size, tumor growth and resistance to treatment. Studies into the expression level of shelterin in oral squamous cell carcinoma (OSCC) report contradictory results. Moreover, the exact role of these proteins in OSCC tumorigenesis remains uncertain. In this review, we examined the data linking telomeres and hallmarks of OSCC. Furthermore, we examined the literature concerning telomeres and the clinical outcome of OSCC. Finally, we propose a model encompassing the role of shelterin proteins in oral tumorigenesis and treatment outcome.


Assuntos
Carcinogênese , Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Telomerase/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Animais , Humanos , Complexo Shelterina , Telômero
4.
Gene Ther ; 22(2): 196-201, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25410741

RESUMO

Simian adeno-associated virus (AAV) serotype rh.10 is a promising gene therapy tool, achieving safe, sustained transgene expression in the nervous system, lung, liver and heart in animal models. To date, preexisting immunity in humans has not been confirmed, though exposure is unexpected. We compared the humoral immune response with serotypes AAVrh.10 and AAV9 in mice, and AAVrh.10, AAV9 and AAV2 in 100 healthy humans. Mice, injected-intravenously, raised significantly more anti-AAV9 than anti-AAVrh.10 IgG (immunoglobulins), and sera demonstrated greater neutralizing capacity, correspondingly. Antibody cross-binding studies in mice showed negligible cross-recognition between AAVrh.10, AAV9 and AAV2. In humans, IgG prevalence against the most common human serotype, AAV2, was 72%; AAV9, 47% and AAVrh.10, a surprising, 59%. Yet, neutralizing-antibody seroprevalences were 71% for AAV2, 18% for AAV9 and 21% for AAVrh.10. Thus, most anti-AAV9 and anti-AAVrh.10 IgG were nonneutralizing. Indeed, sera generally neutralized AAV2 more strongly than AAVrh.10. Further, all samples neutralizing AAVrh.10 or AAV9 also neutralized AAV2, suggesting antibody cross-recognition. This contrasts with the results in mice, and highlights the complexity of tailoring gene therapy to minimize the immune response in humans, when multiple-mixed infections during a lifetime evoke a broad repertoire of preexisting antibodies capable of cross reacting with non-human serotypes.


Assuntos
Anticorpos Antivirais/sangue , Dependovirus/imunologia , Terapia Genética , Animais , Anticorpos Antivirais/imunologia , Reações Cruzadas , Humanos , Masculino , Camundongos Endogâmicos ICR , Ligação Proteica , Transdução Genética
5.
Leukemia ; 29(5): 1163-76, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25394713

RESUMO

Deregulated expression of glycolytic enzymes contributes not only to the increased energy demands of transformed cells but also has non-glycolytic roles in tumors. However, the contribution of these non-glycolytic functions in tumor progression remains poorly defined. Here, we show that elevated expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), but not of other glycolytic enzymes tested, increased aggressiveness and vascularization of non-Hodgkin's lymphoma. Elevated GAPDH expression was found to promote nuclear factor-κB (NF-κB) activation via binding to tumor necrosis factor receptor-associated factor-2 (TRAF2), enhancing the transcription and the activity of hypoxia-inducing factor-1α (HIF-1α). Consistent with this, inactive mutants of GAPDH failed to bind TRAF2, enhance HIF-1 activity or promote lymphomagenesis. Furthermore, elevated expression of gapdh mRNA in biopsies from diffuse large B-cell non-Hodgkin's lymphoma patients correlated with high levels of hif-1α, vegf-a, nfkbia mRNA and CD31 staining. Collectively, these data indicate that deregulated GAPDH expression promotes NF-κB-dependent induction of HIF-1α and has a key role in lymphoma vascularization and aggressiveness.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Gliceraldeído 3-Fosfato Desidrogenase (NADP+)/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Linfoma não Hodgkin/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Animais , Biópsia , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , Células HeLa , Humanos , Linfoma/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Oncogene ; 31(13): 1683-94, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21909141

RESUMO

The anti-VEGF targeted antibody bevacizumab (BVZ) has been approved for treating renal cell carcinomas (RCCs). Although BVZ increases the progression-free survival of patients with metastatic RCC, the effect on overall survival is poor. To gain insight into the limited efficacy of BVZ on overall survival, we analyzed patient samples of RCC for angiogenic factors that may participate in escape from anti-VEGF therapy. Our study shows that the level of vascular endothelial growth factor (VEGF) in tumors was increased compared with normal tissue. The level of interleukin-8/CXCL8, a pro-angiogenic member of the CXCL family of cytokines, was also increased in tumors. These observations gave us a good reason to analyze the combined effects of BVZ and anti-CXCL8 antibodies on tumor growth. Surprisingly, we report that BVZ accelerates the growth of RCC in nude mice with in vivo selection of tumor cells with an increased growth capacity. Downregulation of receptor tyrosine phosphatase-κ, a phosphatase implicated in EGF receptor regulation, may partly explain this phenomenon. Modification of the vascular network and development of lymphatic vessels through VEGF-C production and compensatory production of pro-angiogenic CXCL cytokines were also observed. The apparent normalization of the vascular network prompted us to associate BVZ with the chemotherapeutic agent paclitaxel. While efficient in vitro, paclitaxel did not reverse the anti-VEGF effects in vivo. Anti-CXCL8-targeting antibodies were promising as they decreased intra-tumor VEGF production; decreased the pro-angiogenic CXCL/anti-angiogenic CXCL ratio and did not induce lymphangiogenesis. These observations hold clinical implication as they highlight putative markers implicated in escape from BVZ treatment. They also recommend proceeding with caution in the use of anti-VEGF therapy alone for treatment of RCC.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Interleucina-8/metabolismo , Neoplasias Renais/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bevacizumab , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/patologia , Proliferação de Células , Feminino , Humanos , Interleucina-8/imunologia , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
7.
Int J Cancer ; 130(12): 2771-82, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21805474

RESUMO

Ursodeoxycholic acid (UDCA) attenuates colon carcinogenesis in humans and in animal models by an unknown mechanism. We investigated UDCA effects on normal intestinal epithelium in vivo and in vitro to identify the potential chemopreventive mechanism. Feeding of mice with 0.4% UDCA reduced cell proliferation to 50% and suppressed several potential proproliferatory genes including insulin receptor substrate 1 (Irs-1). A similar transcriptional response was observed in the rat intestinal cell line IEC-6 which was then used as an in vitro model. UDCA slowed down the proliferation of IEC-6 cells and induced sustained hyperphosphorylation of ERK1/ERK2 kinases which completely inhibited the proproliferatory effects of EGF and IGF-1. The hyperphosphorylation of ERK1 led to a transcriptional suppression of the Irs-1 gene. Both, the hyperphosphorylation of ERK as well as the suppression of Irs-1 were sufficient to inhibit proliferation of IEC-6 cells. ERK1/ERK2 inhibition in vitro or ERK1 elimination in vitro or in vivo abrogated the antiproliferatory effects of UDCA. We show that UDCA inhibits proliferation of nontransformed intestinal epithelial cells by inducing a sustained hyperphosphorylation of ERK1 kinase which slows down the cell cycle and reduces expression of Irs-1 protein. These data extend our understanding of the physiological and potentially chemopreventive effects of UDCA and identify new targets for chemoprevention.


Assuntos
Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Ácido Ursodesoxicólico/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Fator de Crescimento Epidérmico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Fator de Crescimento Insulin-Like I/metabolismo , Mucosa Intestinal/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Ácido Ursodesoxicólico/metabolismo
8.
Gene Ther ; 18(6): 622-30, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21326330

RESUMO

Efficient transduction of the peripheral nervous system (PNS) is required for gene therapy of acquired and inherited neuropathies, neuromuscular diseases and for pain treatment. We have characterized the tropism and transduction efficiency of different adeno-associated vectors (AAV) pseudotypes after sciatic nerve injection in the mouse. Among the pseudotypes tested, AAV2/1 transduced both Schwann cells and neurons, AAV2/2 infected only sensory neurons and AAV2/8 preferentially transduced Schwann cells. AAV2/8 expression in the sciatic nerve was detected up to 10 weeks after administration, the latest time point analyzed. The injected mice developed neutralizing antibodies against all AAVs tested; the titers were higher against AAV2/1 than AAV2/2 and were the lowest for AAV2/8, correlating with a higher transgene expression overtime. AAV2/8 coding for ciliary neurotrophic factor (CNTF) led to an upregulation of P0 and PMP22 myelin proteins, four weeks after transduction of injured sciatic nerves. Importantly, CNTF-transduced mice showed a significant increase in both GAP43 expression in sensory neurons, a marker of axonal regeneration, and the compound muscle action potential. These results prove the utility of AAV8 as a gene therapy vector for Schwann cells to treat myelin disorders or to improve nerve regeneration.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Regeneração Nervosa/genética , Animais , Anticorpos Neutralizantes/biossíntese , Linhagem Celular , Fator Neurotrófico Ciliar/metabolismo , Dependovirus/imunologia , Proteína GAP-43/metabolismo , Vetores Genéticos , Injeções , Camundongos , Proteínas da Mielina/metabolismo , Nervos Periféricos , Células de Schwann , Células Receptoras Sensoriais/metabolismo , Sorotipagem , Transdução Genética
9.
Am J Physiol Cell Physiol ; 299(2): C189-202, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20463170

RESUMO

Intracellular signaling by mitogen-activated protein (MAP) kinases (MAPK) is involved in many cellular responses and in the regulation of various physiological and pathological conditions. Tight control of the localization and duration of extracellular-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), or p38 MAPK activity is thus a fundamental aspect of cell biology. Several members of the dual-specificity phosphatase (DUSPs) family are able to dephosphorylate MAPK isoforms with different specificity, cellular, and tissue localization. Understanding how these phosphatases are themselves regulated during development or in physiological and pathological conditions is therefore fundamental. Over the years, gene deletion and knockdown studies have completed initial in vitro studies and shed a new light on the global and specific roles of DUSPs in vivo. Whereas DUSP1, DUSP2, and DUSP10 appear as crucial players in the regulation of immune responses, other members of the family, like the ERK-specific DUSP6, were shown to play a major role in development. Recent findings on the involvement of DUSPs in cancer progression and resistance will also be discussed.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Neoplasias/enzimologia , Neoplasias/etiologia , Animais , Humanos , Isoenzimas/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia
10.
Cell Death Differ ; 17(1): 158-69, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19680261

RESUMO

DJ-1 was recently identified as a gene product responsible for a subset of familial Parkinson's disease (PD). The mechanisms by which mutations in DJ-1 alter its function and account for PD-related pathology remained largely unknown. We show that DJ-1 is processed by caspase-6 and that the caspase-6-derived C-terminal fragment of DJ-1 fully accounts for associated p53-dependent cell death. In line with the above data, we show that a recently described early-onset PD-associated mutation (D149A) renders DJ-1 resistant to caspase-6 proteolysis and abolishes its protective phenotype. Unlike the D149A mutation, the L166P mutation that prevents DJ-1 dimerization does not impair its proteolysis by caspase-6 although it also abolishes DJ-1 antiapoptotic function. Therefore, we show here that DJ-1 loss of function could be due to impaired caspase-6 proteolysis and we document the fact that various DJ-1 mutations could lead to PD pathology through distinct molecular mechanisms.


Assuntos
Caspase 6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Proteínas Oncogênicas/genética , Doença de Parkinson/genética , Substituição de Aminoácidos , Animais , Apoptose , Encéfalo/metabolismo , Células Cultivadas , Dimerização , Regulação para Baixo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas/metabolismo , Doença de Parkinson/metabolismo , Proteína Desglicase DJ-1 , Proteína Supressora de Tumor p53/metabolismo
11.
Acta Physiol (Oxf) ; 196(2): 259-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18983460

RESUMO

AIM: To analyse the correlation between production of angiogenic [vascular endothelial growth factor A (VEGF-A) and interleukin 8 (IL-8)] and lymphangiogenic factors (VEGF-C and D) and adaptation to high altitude (>8000 m). Erythropoietin (EPO) served as a positive control. METHODS: We analysed the percentage of oxygen saturation and the plasmatic contents of VEGF-A, C, D, IL-8 and EPO in seven mountaineers and four Sherpas during an expedition to Mount Everest. Acute mountain sickness was also evaluated using the Lake Louise score. RESULTS: Whereas VEGF-A, IL-8, VEGF-C and EPO were transiently up-regulated at 5000 m and decreased at the highest altitudes, VEGF-D remained elevated throughout the ascent. Sherpas had increased basal levels of VEGF-A, C, IL-8 and EPO and up-regulation of all the tested factors when they passed the altitude at which they lived. CONCLUSION: Our data suggest that expression of angiogenic and lymphangiogenic factors is up-regulated directly or indirectly by altitude-dependent hypoxia. Both factors could be involved in a mechanism of adaptation to high altitudes.


Assuntos
Aclimatação/fisiologia , Altitude , Proteínas Angiogênicas/sangue , Montanhismo , Adulto , Doença da Altitude/diagnóstico , Eritropoetina/sangue , Feminino , Humanos , Hipóxia/sangue , Interleucina-8/sangue , Linfangiogênese/fisiologia , Pessoa de Meia-Idade , Neovascularização Fisiológica/fisiologia , Oxigênio/sangue , Fator A de Crescimento do Endotélio Vascular/sangue , Fator C de Crescimento do Endotélio Vascular/sangue , Fator D de Crescimento do Endotélio Vascular/sangue
12.
Oncogene ; 27(26): 3685-91, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18223677

RESUMO

MAP kinases phosphatases (MKPs) belong to the dual-specificity phosphatase family (DUSP) and dephosphorylate phosphothreonine and phosphotyrosine within MAP kinases. We had previously shown that DUSP6/MKP-3 was phosphorylated and degraded upon growth factor stimulation, in a MEK-dependent manner. Here we show that another pathway involved in growth factor signaling, the PI3K/mTOR signaling pathway, accounts for a part of the phosphorylation and degradation of DUSP6 induced by serum growth factors, as evidenced by experiments using pharmacological inhibitors of PI3 kinase and mammalian target of rapamycin (mTOR). Moreover, specific agonists of the mTOR pathway, such as amino acids or insulin/IGF-1, which do not activate extracellular signal regulated kinases (ERKs) in our cellular model, were also able to induce the phosphorylation and degradation of DUSP6. However, a basal activity of MEK was required for the mTOR pathway-mediated phosphorylation to occur. Mutagenesis studies identified serine 159 within DUSP6 as the target of the mTOR pathway. The ERK phosphatase DUSP6 may thus constitute a novel branch-point of the crosstalk between two major signaling pathways induced by growth factors, the MEK/ERK pathway and the PI3K/mTOR pathway.


Assuntos
Fosfatase 6 de Especificidade Dupla/metabolismo , Proteínas Quinases/fisiologia , Processamento de Proteína Pós-Traducional , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Cricetinae , Fator de Crescimento Insulin-Like I/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Serina-Treonina Quinases TOR
13.
Ann Oncol ; 18(8): 1335-41, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17693647

RESUMO

BACKGROUND: To examine the impact of a frequent her2 gene polymorphism (Ile655Val) on tumor growth and on the pharmacodynamics of treatment by trastuzumab. PATIENTS AND METHODS: Experimental study: The growth characteristics of cells expressing the Ile or Val isoform were examined in vitro and after injection into nude mice. The effect of trastuzumab was determined in both experimental models. Clinical study: 61 patients with advanced breast cancers and treated by trastuzumab were genotyped for HER2 by PCR-RFLP. The influence of HER2 genotype on the trastuzumab treatment was examined. RESULTS: Experimental study: HER2-expressing cells acquired the characteristics of tumor cells. The Val isoform-expressing cells showed the highest growth capacity and developed aggressive tumors sensitive to trastuzumab. Clinical study: There was no link between tumor response or survival and HER2 genotype. All cases of treatment-related cardiotoxicity were found in the Ile/Val group and there was no cardiac toxicity in the Val/Val and Ile/Ile patients. CONCLUSIONS: This study establishes a clear-cut difference between the two HER2 isoforms regarding their tumorogenic potential with an advantage for the Val/HER2 isoform. In breast cancer patients treated with trastuzumab, the presence of a Val allele may constitute a risk factor for cardiac toxicity.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Coração/efeitos dos fármacos , Receptor ErbB-2/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais Humanizados , Sequência de Bases , Western Blotting , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Feminino , Cardiopatias/induzido quimicamente , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição , Isoformas de Proteínas/genética , Transfecção , Trastuzumab
14.
Oncogene ; 25(53): 7096-105, 2006 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-16715126

RESUMO

Transcription factor Sp1 has recently been shown to be overexpressed in a number of human cancers and its overexpression contributes to malignant transformation. Sp1 regulates the expression of a number of genes participating in multiple aspects of tumorigenesis such as angiogenesis, cell growth and apoptosis resistance. To better understand the role of increased Sp1 levels on apoptosis regulation we have used retroviruses to overexpress this protein in haematopoietic Baf-3 cells and in 3T3 fibroblasts. We have also used inducible expression systems to control ectopic Sp1 levels in different cell types. Surprisingly, Sp1 overexpression on its own induces apoptosis in all the cellular models tested. The apoptotic pathways induced by Sp1 overexpression are cell type specific. Finally, using a truncated form of Sp1, we show that Sp1-induced apoptosis requires its DNA-binding domain. Our results highlight that Sp1 levels in untransformed cells must be tightly regulated as Sp1 overexpression leads to the induction of apoptosis. Our results also suggest that cancer cells overexpressing Sp1 can avoid Sp1-induced apoptosis.


Assuntos
Apoptose , Fator de Transcrição Sp1/metabolismo , Animais , DNA , Expressão Gênica , Humanos , Camundongos , Fator de Transcrição Sp1/genética
15.
Br J Cancer ; 94(10): 1516-23, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16641910

RESUMO

Vascular endothelial growth factor-A (VEGF-A) has been demonstrated to play an important role in tumour angiogenesis and to influence prognosis in many cancers. However its prognostic value in head and neck squamous cell carcinomas (HNSCCs) remains controversial. Therefore, we investigated the clinical relevance of VEGF-A expression in HNSCCs and analysed whether its expression was associated with PAIP2 protein levels, a VEGF-A mRNA-binding partner that strongly regulates VEGF-A expression in tissue culture. We determined the correlation of VEGF-A and PAIP2 protein levels, quantitatively evaluated in tumour tissue homogenates from 54 patients with HNSCC, to clinicopathological parameters. We showed that VEGF-A expression in HNSCC is correlated to the stage of tumour differentiation (P=0.050) and is an independent prognostic factor for progression-free survival (P=0.001) and overall survival (P=0.0004). In a pharynx carcinoma cell line, we demonstrated by RNA interference that VEGF-A expression is closely controlled by PAIP2. Moreover, in human HNSCCs, VEGF-A expression is significantly correlated to PAIP2 protein levels (P=0.0018). Nevertheless, PAIP2 expression is associated with neither clinicopathological factors nor patient's survival. Our data suggest that, in contrast to PAIP2 protein levels, which are unrelated to tumour prognosis, VEGF-A expression could serve as a prognostic marker in head and neck cancer and may be helpful for targeted therapies.


Assuntos
Neoplasias de Cabeça e Pescoço/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Northern Blotting , Western Blotting , Diferenciação Celular , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Neoplasias Laríngeas , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética , Estudos Retrospectivos , Taxa de Sobrevida , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética
16.
Novartis Found Symp ; 240: 186-96; discussion 196-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11727929

RESUMO

Signalling via growth factors, oncogenes and environmental stresses such as hypoxia, promotes the up-regulation of glycolysis, intracellular pH (pHi) and vascular endothelial growth factor (VEGF) via cooperative mechanisms. Somatic cell genetics was applied to a fibroblastic cell line (CCOL39) to disrupt either aerobic glycolysis, respiration, or a major pHi-regulating system, the Na-H exchanger (NHE1). We obtained stable mutants impaired either in phosphoglucose isomerase (pgi-), which produce virtually no lactic acid, or in respiration (res-), which over secrete lactic acid (three- to fourfold the wild-type rate). These mutations, which allowed us to analyse the incidence of lactic acid production on tumour development in nude mice, were analysed alone, or in combination, with the mutation nhe1- to evaluate in vivo the role of NHE1 on pHi control and cell proliferation. Ras-transformed pgi- cells (not forming lactic acid) form tumours like wild type transformed cells (100% incidence). The disruption of NHE1 however, strongly reduced tumour incidence to about 20%. In cells bearing both mutations, nhe1-, res-, and which therefore over-produce lactic acid, the situation is even more dramatic (0% incidence). In sharp contrast, association of nhe1- with pgi- restored 100% tumour incidence. We conclude that over-production of lactic acid is detrimental for tumour development and that NHE1, by controlling pHi, plays a key role in cell survival/proliferation and tumour growth. Finally we summarize our current knowledge on the signalling mechanisms leading to VEGF expression, another key component of tumour growth via neo-vascularization.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Glicólise , Concentração de Íons de Hidrogênio , Linfocinas/fisiologia , Neoplasias/patologia , Aerobiose , Animais , Divisão Celular , Humanos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Biotechniques ; 30(6): 1254-6, 1258-60, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11414215

RESUMO

The vascular endothelial growth factor (VEGF) is implicated in the progression of cancers. Its expression is well correlated with tumor growth and metastases. The availability of a rapid and sensitive method to detect the amounts of VEGF mRNA in biological samples of limited size, very small biopsies, or samples containing relatively few cells could provide an interesting prognostic tool for clinicians. We have developed an RT-PCR method that allows us to detect the VEGF mRNA from as little as 3 micrograms total mRNA. We have also shown that this protocol can be generalized to all cell lines tested. This method constitutes a very potent tool for the analysis of VEGF mRNA expression in different contexts.


Assuntos
Fatores de Crescimento Endotelial/biossíntese , Linfocinas/biossíntese , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adenocarcinoma/patologia , Animais , Ligação Competitiva , Biópsia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , Neoplasias do Colo/patologia , Cricetinae , Primers do DNA/química , Primers do DNA/genética , DNA Complementar/genética , Fatores de Crescimento Endotelial/genética , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Linfocinas/genética , Camundongos , Microquímica/métodos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/genética , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/análise , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Ratos , Padrões de Referência , Deleção de Sequência , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Biochem Pharmacol ; 60(8): 1171-8, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11007955

RESUMO

Angiogenesis is associated with a number of pathological situations. In this study, we have focused our attention on the role of p42/p44 MAP (mitogen-activated protein) kinases and hypoxia in the control of angiogenesis. We demonstrate that p42/p44 MAP kinases play a pivotal role in angiogenesis by exerting a determinant action at three levels: i) persistent activation of p42/p44 MAP kinases abrogates apoptosis; ii) p42/p44 MAP kinase activity is critical for controlling proliferation and growth arrest of confluent endothelial cells; and iii) p42/p44 MAP kinases promote VEGF (vascular endothelial growth factor) expression by activating its transcription via recruitment of the AP-2/Sp1 (activator protein-2) complex on the proximal region (-88/-66) of the VEGF promoter and by direct phosphorylation of hypoxia-inducible factor 1 alpha (HIF-1 alpha). HIF-1 alpha plays a crucial role in the control of HIF-1 activity, which mediates hypoxia-induced VEGF expression. We show that oxygen-regulated HIF-1 alpha protein levels are not affected by intracellular localization (nucleus versus cytoplasm). Finally, we propose a model which suggests an autoregulatory feedback mechanism controlling HIF-1 alpha and therefore HIF-1-dependent gene expression.


Assuntos
Hipóxia/fisiopatologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neovascularização Patológica/fisiopatologia , Oxigênio/metabolismo , Fatores de Transcrição , Animais , Sobrevivência Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Endotélio/enzimologia , Expressão Gênica/fisiologia , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Linfocinas/biossíntese , Linfocinas/genética , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Ann N Y Acad Sci ; 902: 187-200, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10865838

RESUMO

Vascular endothelial growth factor (VEGF), a potent agonist secreted by virtually all cells, controls migration and division of vascular endothelial cells. Disruption of one VEGF allele in mice has revealed a dramatic lethal effect in early embryogenesis, suggesting a key role in vasculogenesis. We analyzed the regulation of VEGF mRNA in normal and transformed CCL39 fibroblasts and then dissected the VEGF promoter to identify the signaling pathway(s) controlling the activation of this promoter in response to growth factors, oncogenes, and hypoxic stress. We demonstrated that the p42/p44 MAP kinase signaling cascade controls VEGF expression at least at two levels. In normoxic conditions, MAPKs activate the VEGF promoter at the proximal (-88/-66) region where Sp-1/AP-2 factors bind. Activation of p42/p44 MAPKs is sufficient to turn on VEGF mRNA. At low O2 tension, hypoxia inducible factor-1 alpha (HIF-1 alpha), a limiting factor rapidly stabilized and phosphorylated, plays a key role in the expression of several genes including VEGF. We demonstrated that p42/p44MAPKs stoichiometrically phosphorylate HIF-1 alpha in vitro and that HIF-1-dependent VEGF gene expression is strongly enhanced by the exclusive activation of p42/p44MAPKs. Finally, we demonstrated that the regulation of p42/p44MAPK activity is critical for controlling proliferation and growth arrest of vascular endothelial cells at confluency. These results point to at least three major targets of angiogenesis where p42/p44 MAP kinases exert a determinant action.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Animais , Fatores de Crescimento Endotelial/genética , Humanos , Linfocinas/genética , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Oncogenes , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
J Biol Chem ; 275(34): 26484-91, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-10849421

RESUMO

Stability of the vascular endothelial growth factor (VEGF) mRNA is tightly regulated through its 3'-untranslated region (3'-UTR). Here, we demonstrate that VEGF mRNA levels are increased by anisomycin, a strong activator of stress-activated protein kinases. Hence, VEGF mRNA induction is inhibited by SB202190, an inhibitor of JNK and p38/HOG kinase. Furthermore, VEGF mRNA expression is increased in cells that overexpress JNK and p38/HOG by an increase in its stability. We show by two different approaches that anisomycin exerts its effect on the VEGF mRNA 3'-UTR. First, by using an in vitro mRNA degradation assay, the half-life of the VEGF mRNA 3'-UTR region transcript was found to be increased when incubated with extracts from anisomycin-treated cells; and second, the 3'-UTR was also sufficient to confer mRNA instability to the Nhe3 (Na(+)/H(+) exchanger 3) heterologous reporter gene, and anisomycin treatment stabilized the chimeric mRNA (Nhe3 fused to the VEGF mRNA 3'-UTR). This chimeric mRNA is also more stable in cells overexpressing p38/HOG and JNK that have been stimulated by anisomycin. We show that such regulation is mediated through an AU-rich region of the 3'-UTR contained within a stable hairpin structure. By RNA electrophoretic mobility shift assays, we show that this region binds proteins specifically induced by anisomycin treatment. These findings clearly demonstrate a major role of stress-activated protein kinases in the post-transcriptional regulation of VEGF.


Assuntos
Fatores de Crescimento Endotelial/genética , Linfocinas/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Saccharomyces cerevisiae , Anisomicina/farmacologia , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Conformação de Ácido Nucleico , Inibidores da Síntese de Proteínas/farmacologia , Piridinas/farmacologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA