Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Haematol ; 192(2): 385-394, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33169863

RESUMO

Previously, we showed that nearly 70% of children followed in our sickle cell disease (SCD) clinic were vitamin D- deficient and had low vitamin intake with poor use of supplements. We compared the change in serum 25-hydroxyvitamin D [25(OH)D], safety and clinical impact of two vitamin D supplementation regimens in children with SCD. Children (5-17 years, all genotypes) were randomized to a single bolus of vitamin D3 (300 000 IU; n = 18) or placebo (n = 20). All children received a prescription for daily 1 000 IU vitamin D3 . Serum 25(OH)D and calcium, urinary calcium/creatinine ratio, musculoskeletal pain, quality of life, haematology and bone markers were assessed at baseline and three months post intervention. Bolus administration led to a greater rise in 25(OH)D levels from baseline compared to placebo (20 ± 16 nmol/l vs. 2 ± 19 nmol/l; P = 0·003) and correction of vitamin D deficiency. No hypercalcaemia nor hypercalciuria occurred during the study, but more children in the bolus group experienced gastrointestinal symptoms within the first month (P = 0·04). There were no differences between groups for other outcomes. The use of a high-dose vitamin D bolus combined with daily 1 000 IU vitamin D3 was more efficient in raising 25(OH)D levels than daily supplementation alone in children with SCD.


Assuntos
Anemia Falciforme/tratamento farmacológico , Colecalciferol/uso terapêutico , Deficiência de Vitamina D/tratamento farmacológico , Vitaminas/uso terapêutico , Adolescente , Anemia Falciforme/sangue , Anemia Falciforme/complicações , Cálcio/sangue , Criança , Pré-Escolar , Colecalciferol/administração & dosagem , Suplementos Nutricionais , Feminino , Humanos , Masculino , Resultado do Tratamento , Vitamina D/análogos & derivados , Vitamina D/sangue , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/complicações , Vitaminas/administração & dosagem
2.
BMC Cancer ; 19(1): 1018, 2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664947

RESUMO

BACKGROUND: Neuroblastoma (NB) is a frequent pediatric tumor associated with poor prognosis. The disregulation of Bcl-2, an anti-apoptotic protein, is crucial for the tumoral development and chemoresistance. Autophagy is also implicated in tumor cell survival and chemoresistance. The aim of our study was to demonstrate therapeutic efficiency of GX 15-070, a pan-Bcl-2 family inhibitor, used alone and in combination with conventional drugs or with hydroxychloroquine (HCQ), an autophagy inhibitor. METHODS: Five neuroblastoma cell lines were tested for the cytotoxic activity of GX 15-070 alone or in combination with cisplatin, doxorubicin, HCQ or Z-VAD-FMK a broad-spectrum caspase inhibitor. Apoptosis and autophagy levels were studied by western-blot and FACS. Orthotopic injections were performed on NOD/LtSz-scid/IL-2Rgamma null mice that were treated with either GX 15-070 alone or in combination with HCQ. RESULTS: Synergistic cytotoxicity was observed for the drug combination in all of the 5 neuroblastoma cell lines tested, including MYCN amplified lines and in cancer stem cells. GX 15-070 significantly increased apoptosis and autophagy in neuroblastoma cells as evidenced by increased levels of the autophagy marker, LC3-II. Inhibition of autophagy by HCQ, further increased the cytotoxicity of this combinatorial treatment, suggesting that autophagy induced by these agent plays a cytoprotective role. In vivo, GX 15-070 combined with HCQ significantly decreased the growth of the tumor and the number of distant metastases. CONCLUSIONS: Based on the synergistic effect of HCQ and GX 15-070 observed in this study, the combination of these two drugs may be utilized as a new therapeutic approach for neuroblastoma.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Pirróis/farmacologia , Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Hidroxicloroquina/farmacologia , Hidroxicloroquina/uso terapêutico , Indóis , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Pirróis/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Artigo em Inglês | MEDLINE | ID: mdl-31568924

RESUMO

Nutritional deficiencies often precede the diagnosis of cystic fibrosis (CF) in infants, and occur at a stage where the rapidly developing brain is more vulnerable to insult. We aim to compare fat-soluble nutrient status of newly diagnosed non-screened infants with CF to that of healthy infants, and explore the association with neurodevelopment evaluated by electroencephalography (EEG). Our results show that CF infants had lower levels of all fat-soluble vitamins and docosahexaenoic acid (DHA) compared to controls. The auditory evoked potential responses were higher in CF compared to controls whereas the visual components did not differ between groups. DHA levels were correlated with auditory evoked potential responses. Although resting state frequency power was similar between groups, we observed a negative correlation between DHA levels and low frequencies. This study emphasizes the need for long-term neurodevelopmental follow-up of CF infants and pursuing intervention strategies in the future.


Assuntos
Fibrose Cística/fisiopatologia , Ácidos Docosa-Hexaenoicos/análise , Potenciais Evocados Auditivos , Potenciais Evocados Visuais , Vitaminas/análise , Estudos de Casos e Controles , Fibrose Cística/metabolismo , Eletroencefalografia , Feminino , Humanos , Lactente , Masculino , Projetos Piloto , Descanso
4.
BMC Cancer ; 16(1): 891, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27846885

RESUMO

BACKGROUND: Neuroblastoma (NB) is a frequent pediatric tumor characterized by a poor prognosis where a majority of tumors progress despite intensive multimodality treatments. Autophagy, a self-degradative process in cells, could be induced by chemotherapy and be associated with chemoresistance. The aim of this study was to determine whether: 1) autophagy is present in NB, 2) chemotherapy modified its levels, and 3) its inhibition decreased chemoresistance. METHODS: Immunohistochemical stainings were performed on samples from 184 NB patients in order to verify the expression of LC3B, a specific marker for autophagy, and Beclin 1, a positive regulator of autophagy. In addition, we performed an in vitro study with six NB cell lines and six drugs (vincristine, doxorubicin, cisplatin temozolomide, LY294002 and syrolimus). Inhibition of autophagy was performed using ATG5 knockdown cells or hydroxychloroquine (HCQ). Cell survival was measured using the MTT cell proliferation assay. Autophagy was detected by monodansylcadaverine, confocal microscopy and Western blot. In vivo study with tumor xenografts in NSG mice was performed. RESULTS: Our results have indicated that autophagy was present at low levels in NB and was not a prognostic factor, while Beclin 1 was highly expressed in children with poor NB prognosis. However, autophagy levels increased after chemotherapy in vitro and in vivo. Tumor progression was significantly decreased in mice treated with a combination of HCQ and vincristine. CONCLUSIONS: Taken together, autophagy is present in NB, induced by chemotherapy and associated with chemoresistance, which is significantly reduced by its inhibition. Therefore, targeting autophagy represents a very attractive approach to develop new therapeutic strategies in NB.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Animais , Antineoplásicos/uso terapêutico , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células , Criança , Pré-Escolar , Modelos Animais de Doenças , Progressão da Doença , Seguimentos , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Camundongos , Camundongos Knockout , Estadiamento de Neoplasias , Neuroblastoma/diagnóstico , Neuroblastoma/tratamento farmacológico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
5.
PLoS One ; 7(8): e43665, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916293

RESUMO

Neuroblastoma (NB) is a typical childhood and heterogeneous neoplasm for which efficient targeted therapies for high-risk tumors are not yet identified. The chemokine CXCL12, and its receptors CXCR4 and CXCR7 have been involved in tumor progression and dissemination. While CXCR4 expression is associated to undifferentiated tumors and poor prognosis, the role of CXCR7, the recently identified second CXCL12 receptor, has not yet been elucidated in NB. In this report, CXCR7 and CXCL12 expressions were evaluated using a tissue micro-array including 156 primary and 56 metastatic NB tissues. CXCL12 was found to be highly associated to NB vascular and stromal structures. In contrast to CXCR4, CXCR7 expression was low in undifferentiated tumors, while its expression was stronger in matured tissues and specifically associated to differentiated neural tumor cells. As determined by RT-PCR, CXCR7 expression was mainly detected in N-and S-type NB cell lines, and was slightly induced upon NB cell differentiation in vitro. The relative roles of the two CXCL12 receptors were further assessed by overexpressing CXCR7 or CXCR4 receptor alone, or in combination, in the IGR-NB8 and the SH-SY5Y NB cell lines. In vitro functional analyses indicated that, in response to their common ligand, both receptors induced activation of ERK1/2 cascade, but not Akt pathway. CXCR7 strongly reduced in vitro growth, in contrast to CXCR4, and impaired CXCR4/CXCL12-mediated chemotaxis. Subcutaneous implantation of CXCR7-expressing NB cells showed that CXCR7 also significantly reduced in vivo growth. Moreover, CXCR7 affected CXCR4-mediated orthotopic growth in a CXCL12-producing environment. In such model, CXCR7, in association with CXCR4, did not induce NB cell metastatic dissemination. In conclusion, the CXCR7 and CXCR4 receptors revealed specific expression patterns and distinct functional roles in NB. Our data suggest that CXCR7 elicits anti-tumorigenic functions, and may act as a regulator of CXCR4/CXCL12-mediated signaling in NB.


Assuntos
Quimiocina CXCL12/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Movimento Celular , Quimiocina CXCL12/genética , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Técnicas In Vitro , Neuroblastoma/genética , Fosforilação , Receptores CXCR/genética , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
6.
Genes Chromosomes Cancer ; 51(8): 792-804, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22514116

RESUMO

Neuroblastoma (NB) is the most common and lethal extracranial solid tumor of childhood. Despite aggressive therapy, more than half of the children with advanced NB will die of uncontrolled metastatic disease. After chemotherapy, tumor-initiating cells (TICs) could persist, cause relapses and metastasis. The aim of this study is to demonstrate the tumor-initiating properties of CD133high NB cells and to identify new specific genetic abnormalities. Isolation of the CD133high cell population from NB cell lines was followed by neurosphere formation, soft agar assays, and orthotopic injections in NOD/SCID/IL2Rγc-null mice. A differential genotyping analysis was performed with Affymetrix SNP 6.0 arrays on CD133low and CD133high populations and the frequency of the abnormalities of 36 NB tumors was determined. Our results show that CD133high NB cells possess tumor-initiating properties, as CD133high cells formed significantly more neurospheres and produced significantly more colonies in soft agar than CD133low. Injection of 500 CD133high cells was sufficient to generate primary tumors and frequent metastases in mice. Differential genotyping analysis demonstrated two common regions with gains (16p13.3 and 19p13.3) including the gene EFNA2 in the CD133high population, and two with loss of heterozygosity (16q12.1 and 21q21.3) in the CD133low population. The gain of EFNA2 correlated with increased expression of the corresponding protein. These abnormalities were found in NB samples and some were significantly correlated with CD133 expression. Our results show that CD133high NB cells have TICs properties and present different genotyping characteristics compared to CD133low cells. Our findings reveal insights into new therapeutic targets in NB TICs.


Assuntos
Antígenos CD/genética , Glicoproteínas/genética , Neuroblastoma/genética , Peptídeos/genética , Antígeno AC133 , Neoplasias das Glândulas Suprarrenais , Animais , Antígenos CD/biossíntese , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Separação Celular , Distribuição de Qui-Quadrado , Aberrações Cromossômicas , Feminino , Genótipo , Glicoproteínas/biossíntese , Glicoproteínas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutagênese Insercional , Transplante de Neoplasias , Neuroblastoma/metabolismo , Peptídeos/metabolismo , Polimorfismo de Nucleotídeo Único
7.
Histopathology ; 60(7): 1144-55, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22394107

RESUMO

AIMS: Neuroblastoma is a frequent childhood cancer with a heterogeneous prognosis. CD133 expression is an independent prognostic marker for a low survival rate in several cancers. The aim of this study was to determine the prognostic value of CD133 expression in a large cohort of neuroblastoma cases, to define the chemoresistance of neuroblastoma cells expressing CD133, and to determine whether this chemoresistance is regulated by activation of the AKT pathway. METHODS AND RESULTS: Two hundred and eighty samples of neuroblastoma were screened for CD133 expression. The sensitivity of purified CD133+ neuroblastoma cells isolated from two human cell lines to doxorubicin, vincristine and cisplatin, as single agents or in combination with LY294002, an AKT inhibitor, was evaluated in vitro. CD133 expression was found in 100 of 280 tumours. There was a significant association between CD133 expression and the following poor prognosis covariates: age, International Neuroblastoma Staging System stage, MYCN amplification, and phospho-AKT (pAKT) expression. Patients with CD133- tumours had significantly better 3-year event-free and overall survival than patients with CD133+ tumours. In a multivariate model, CD133 expression was independently associated with decreased overall survival. CD133(high) neuroblastoma cells were significantly resistant to chemotherapy as compared with CD133(low) cells. Treatment of unsorted neuroblastoma cells with the three anticancer drugs significantly enriched the CD133+ subpopulation. CD133(high) cells expressed significantly higher levels of pAKT than CD133(low) cells. LY294002 treatment abolished the preferential survival of CD133(high) cells. CONCLUSIONS: CD133 is associated with in-vitro resistance to chemotherapy involving activation of the AKT pathway.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Neuroblastoma/tratamento farmacológico , Neuroblastoma/imunologia , Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antígeno AC133 , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Linhagem Celular Tumoral , Criança , Pré-Escolar , Cromonas/administração & dosagem , Cisplatino/administração & dosagem , Estudos de Coortes , Doxorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Amplificação de Genes , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Morfolinas/administração & dosagem , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Prognóstico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais , Vincristina/administração & dosagem
8.
In Vivo ; 26(1): 19-29, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22210712

RESUMO

In order to develop a relevant xenogenic animal model of neuroblastoma (NB), we compared the tumorigenicity and metastatic potential of SK-N-SH and SK-N-DZ NB cell lines in nude mice and NOD/SCID Il2rg null (NSG) mice. Subcutaneous injection of cell lines induced tumor formation only in NSG mice and was accompanied by metastasis to the liver, adrenal glands, skull and bone marrow. NSG mice injected intravenously showed a profile of distant metastasis that was not observed in nude mice. In addition, tumor growth rates and organ infiltration patterns associated with injected NB cell lines correlated with the in vitro proliferation properties and genetic markers of poor prognosis in NB patients. We also showed that cisplatin chemotherapy was able to inhibit tumor growth. These results clearly demonstrate the higher tumorigenic and metastatic potential of NB cells in NSG mice. Therefore, this xenograft NB model should prove useful in testing the efficacy of new therapeutic approaches for NB.


Assuntos
Neoplasias das Glândulas Suprarrenais/secundário , Modelos Animais de Doenças , Neoplasias Hepáticas/secundário , Neuroblastoma/patologia , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Antineoplásicos/farmacologia , Antígeno CD56/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Neuroblastoma/genética , Neuroblastoma/metabolismo , Fosfopiruvato Hidratase/metabolismo , Tetraspanina 28/metabolismo , Transplante Heterólogo , Carga Tumoral/efeitos dos fármacos
9.
Anticancer Res ; 30(6): 1887-95, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20651331

RESUMO

We recently reported that membrane-type 1 matrix metalloproteinase (MT1-MMP) is phosphorylated on its unique cytoplasmic tyrosine residue but the contribution of this event to tumor progression remains unclear. In this work, we show that the non phosphorylizable cell-permeable peptide antennapedia-coupled cytoplasmic MMP-14 (ACM-14), consisting of the mutated (Y573F) cytoplasmic domain of MT1-MMP coupled to antennapedia, inhibits tyrosine phosphorylation of the enzyme and markedly reduces tumor cell proliferation within 3D type I collagen matrices. Interestingly, administration of ACM-14 to mice markedly delays tumor progression and increases survival, these antitumor actions being associated with the induction of extensive tumor necrosis. Overall, these findings suggest that inhibition of MT1-MMP tyrosine phosphorylation may represent an attractive strategy for the development of novel anticancer drugs.


Assuntos
Antineoplásicos/farmacologia , Biotina/análogos & derivados , Metaloproteinase 14 da Matriz/metabolismo , Peptídeos/farmacologia , Tirosina/metabolismo , Sequência de Aminoácidos , Animais , Biotina/farmacologia , Caspases/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Feminino , Camundongos , Dados de Sequência Molecular , Fosforilação , Sarcoma Experimental/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
BMC Cancer ; 9: 422, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19961596

RESUMO

BACKGROUND: Neuroblastoma is a pediatric tumor of neural crest cells that is clinically characterized by its variable evolution, from spontaneous regression to malignancy. Despite many advances in neuroblastoma research, 60% of neuroblastoma, which are essentially metastatic cases, are associated with poor clinical outcome due to the lack of effectiveness of current therapeutic strategies. Membrane-type 1 matrix metalloproteinase (MT1-MMP, MMP-14), an enzyme involved in several steps in tumor progression, has previously been shown to be associated with poor clinical outcome for neuroblastoma. Based on our recent demonstration that MT1-MMP phosphorylation is involved in the growth of fibrosarcoma tumors, we examined the potential role of phosphorylated MT1-MMP in neuroblastoma progression. METHODS: Tyrosine phosphorylated MT1-MMP was immunostained on tissue microarray samples from 55 patients with neuroblastoma detected by mass screening (known to be predominantly associated with favourable outcome), and from 234 patients with standard diagnosed neuroblastoma. In addition, the effects of a non phosphorylable version of MT1-MMP on neuroblastoma cell migration and proliferation were investigated within three-dimensional collagen matrices. RESULTS: Although there is no correlation between the extent of tyrosine phosphorylation of MT1-MMP (pMT1-MMP) and MYCN amplification or clinical stage, we observed greater phosphorylation of pMT1-MMP in standard neuroblastoma, while it is less evident in neuroblastoma from mass screening samples (P = 0.0006) or in neuroblastoma samples from patients younger than one year (P = 0.0002). In vitro experiments showed that overexpression of a non-phosphorylable version of MT1-MMP reduced MT1-MMP-mediated neuroblastoma cell migration and proliferation within a three-dimensional type I collagen matrix, suggesting a role for the phosphorylated enzyme in the invasive properties of neuroblastoma cells. CONCLUSION: Overall, these results suggest that tyrosine phosphorylated MT1-MMP plays an important role in neuroblastoma progression and that its expression is preferentially observed in tumor specimens from neuroblastoma patients showing poor clinical outcome.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Biomarcadores Tumorais/análise , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Pré-Escolar , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Masculino , Microscopia Confocal , Fosforilação , Análise Serial de Tecidos , Tirosina/metabolismo
11.
Carcinogenesis ; 29(8): 1655-64, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18621744

RESUMO

Pericellular proteolysis of the extracellular matrix by membrane type 1-matrix metalloproteinase (MT1-MMP) confers tumor cells with the ability to proliferate within three-dimensional (3D) matrices and sustains tumor growth in mice. In this study, we show that in addition to its matrix-degrading activity, phosphorylation of MT1-MMP on its unique tyrosine residue located within its cytoplasmic sequence (Tyr573) may also participate to these processes. Fibrosarcoma cells expressing a proteolytically active but non-phosphorylable mutant of MT1-MMP showed a markedly reduced proliferation rate when embedded within 3D type I collagen matrices, this antiproliferative effect being correlated with arrest in the G(0)/G(1) phase of the cell cycle. Impaired tyrosine phosphorylation of MT1-MMP also inhibits anchorage-independent growth of HT-1080 cells in soft agar as well as their invasion of collagen barriers, two prominent attributes of tumor cells, suggesting a broad inhibitory effect of the MT1-MMP mutant on tumorigenesis. Accordingly, whereas HT-1080 cells formed well-vascularized tumors containing tyrosine-phosphorylated MT1-MMP, tumor growth was completely abolished by expression of the non-phosphorylable MT1-MMP mutant. These findings thus indicate a close co-operation between the matrix-degrading activity of MT1-MMP and tyrosine phosphorylation of its intracellular domain for tumor cell invasion and proliferation and suggest that the targeting of the intracellular signaling pathways leading to tyrosine phosphorylation of MT1-MMP may represent an unexpected alternative strategy for the inhibition of this enzyme.


Assuntos
Fibrossarcoma/patologia , Metaloproteinase 1 da Matriz/metabolismo , Tirosina/metabolismo , Aneuploidia , Animais , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos , Invasividade Neoplásica , Fosforilação , Fosfotirosina , Ratos
12.
FEBS Lett ; 582(3): 399-404, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18164686

RESUMO

Membrane-type 1 matrix metalloproteinase (MT1-MMP) plays an important role in sphingosine-1-phosphate(S1P)-dependent migration of endothelial cells but the underlying mechanisms remain largely unknown. Herein, we show that S1P promotes the relocalization of MT1-MMP to peripheral actin-rich membrane ruffles that is coincident with its association with the adaptor protein p130Cas at the leading edge of migrating cells. Immunoprecipitation and confocal microscopy analyses suggest that this interaction required the tyrosine phosphorylation of p130Cas and also involves S1P-dependent phosphorylation of MT1-MMP within its cytoplasmic sequence. The interaction of MT1-MMP with p130Cas at the cell periphery suggests the existence of a close interplay between pericellular proteolysis and signaling pathways involved in EC migration.


Assuntos
Proteína Substrato Associada a Crk/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Lisofosfolipídeos/farmacologia , Metaloproteinase 14 da Matriz/metabolismo , Esfingosina/análogos & derivados , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Humanos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Tirosina Quinases/metabolismo , Esfingosina/farmacologia , Distribuição Tecidual
13.
Mol Cancer Res ; 5(6): 569-83, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17541067

RESUMO

Proteolysis of extracellular matrix proteins by membrane-type 1 matrix metalloproteinase (MT1-MMP) plays a pivotal role in tumor and endothelial cell migration. In addition to its proteolytic activity, several studies indicate that the proinvasive properties of MT1-MMP also involve its short cytoplasmic domain, but the specific mechanisms mediating this function have yet to be fully elucidated. Having previously shown that the serum factor sphingosine 1-phosphate stimulates MT1-MMP promigratory function through a process that involves its cytoplasmic domain, we now extend these findings to show that this cooperative interaction is permissive to cellular migration through MT1-MMP-dependent transactivation of the epidermal growth factor receptor (EGFR). In the presence of sphingosine 1-phosphate, MT1-MMP stimulates EGFR transactivation through a process that is dependent upon the cytoplasmic domain of the enzyme but not its catalytic activity. The MT1-MMP-induced EGFR transactivation also involves G(i) protein signaling and Src activities and leads to enhanced cellular migration through downstream extracellular signal-regulated kinase activation. The present study, thus, elucidates a novel role of MT1-MMP in signaling events mediating EGFR transactivation and provides the first evidence of a crucial role of this receptor activity in MT1-MMP promigratory function. Taken together, our results suggest that the inhibition of EGFR may represent a novel target to inhibit MT1-MMP-dependent processes associated with tumor cell invasion and angiogenesis.


Assuntos
Receptores ErbB/metabolismo , Metaloproteinase 14 da Matriz/fisiologia , Ativação Transcricional , Animais , Células COS , Bovinos , Diferenciação Celular , Movimento Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Lisofosfolipídeos/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Modelos Biológicos , Neovascularização Patológica , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo
14.
J Biol Chem ; 282(21): 15690-9, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17389600

RESUMO

Membrane type 1 matrix metalloproteinase (MT1-MMP) is a transmembrane MMP that plays important roles in migratory processes underlying tumor invasion and angiogenesis. In addition to its matrix degrading activity, MT1-MMP also contains a short cytoplasmic domain whose involvement in cell locomotion seems important but remains poorly understood. In this study, we show that MT1-MMP is phosphorylated on the unique tyrosine residue located within this cytoplasmic sequence (Tyr(573)) and that this phosphorylation requires the kinase Src. Using phosphospecific antibodies recognizing MT1-MMP phosphorylated on Tyr(573), we observed that tyrosine phosphorylation of the enzyme is rapidly induced upon stimulation of tumor and endothelial cells with the platelet-derived chemoattractant sphingosine-1-phosphate, suggesting a role in migration triggered by this lysophospholipid. Accordingly, overexpression of a nonphosphorylable MT1-MMP mutant (Y573F) blocked sphingosine-1-phosphate-induced migration of Human umbilical vein endothelial cells and HT-1080 (human fibrosarcoma) cells and failed to stimulate migration of cells lacking the enzyme (bovine aortic endothelial cells). Altogether, these findings strongly suggest that the Src-dependent tyrosine phosphorylation of MT1-MMP plays a key role in cell migration and further emphasize the importance of the cytoplasmic domain of the enzyme in this process.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Endoteliais/enzimologia , Lisofosfolipídeos/farmacologia , Metaloproteinase 14 da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Neovascularização Patológica/enzimologia , Esfingosina/análogos & derivados , Quinases da Família src/metabolismo , Substituição de Aminoácidos , Animais , Aorta/enzimologia , Aorta/patologia , Células COS , Bovinos , Linhagem Celular Tumoral , Chlorocebus aethiops , Células Endoteliais/patologia , Humanos , Lisofosfolipídeos/metabolismo , Metaloproteinase 14 da Matriz/genética , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Estrutura Terciária de Proteína/genética , Esfingosina/metabolismo , Esfingosina/farmacologia , Veias Umbilicais/enzimologia , Veias Umbilicais/patologia
15.
J Biol Chem ; 279(50): 52132-40, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15466865

RESUMO

We have recently shown that stimulation of endothelial cells with vascular endothelial growth factor (VEGF) induces dissociation of caveolin-1 from the VEGFR-2 receptor, followed by Src family kinase-dependent tyrosine phosphorylation of the protein (Labrecque, L., Royal, I., Surprenant, D. S., Patterson, C., Gingras, D., and Beliveau, R. (2003) Mol. Biol. Cell 14, 334-347). In this study, we provide evidence that the VEGF-dependent tyrosine phosphorylation of caveolin-1 induces interaction of the protein with the membrane-type 1 matrix metalloproteinase (MT1-MMP). This interaction requires the phosphorylation of caveolin-1 on tyrosine 14 by members of the Src family of protein kinases, such as Src and Fyn, because it is completely abolished by expression of a catalytically inactive Src mutant or by site-directed mutagenesis of tyrosine 14 of caveolin-1. Most interestingly, the association of MT1-MMP with phosphorylated caveolin-1 induced the recruitment of Src and a concomitant inhibition of the kinase activity of the enzyme, suggesting that this complex may be involved in the negative regulation of Src activity. The association of MT1-MMP with phosphorylated caveolin-1 occurs in caveolae membranes and involves the cytoplasmic domain of MT1-MMP because it was markedly reduced by mutation of Cys574 and Val582 residues of the cytoplasmic tail of the enzyme. Most interestingly, the reduction of the interaction between MT1-MMP and caveolin-1 by using these mutants also decreases MT1-MMP-dependent cell locomotion. Overall these results indicate that MT1-MMP associates with tyrosine-phosphorylated caveolin-1 and that this complex may play an important role in MT1-MMP regulation and function.


Assuntos
Caveolinas/química , Caveolinas/metabolismo , Metaloendopeptidases/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Bovinos , Cavéolas/metabolismo , Caveolina 1 , Caveolinas/genética , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Técnicas In Vitro , Metaloproteinases da Matriz Associadas à Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tirosina/química , Fator A de Crescimento do Endotélio Vascular/farmacologia
16.
Biochem Biophys Res Commun ; 320(1): 205-12, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15207722

RESUMO

We recently reported that Neovastat, an antiangiogenic drug that is currently undergoing Phase III clinical trials for the treatment of non-small cell lung cancer, may inhibit angiogenesis through an increase in tPA activity. Here, we show that Neovastat also stimulates tPA gene transcription in endothelial cells, in a TNFalpha-like manner. RT-PCR analysis and gene reporter assays using the human tPA promoter indicated that upregulation of the tPA gene transcription by both Neovastat and TNFalpha was correlated with the phosphorylation of JNK1/2 and of IkappaB and that SP600125 and BAY11-7082, inhibitors of JNK and IkappaK, respectively, inhibit the increase of tPA gene transcription induced by Neovastat and TNFalpha. These results suggest that Neovastat induces tPA gene transcription through activation of the JNK and NFkappaB signaling pathways, leading to an increase of tPA secretion by endothelial cells. This may lead to the localized destruction of the fibrin provisional matrix that is necessary for neovessel formation and thus contribute to the reported antiangiogenic properties of this compound.


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Extratos de Tecidos/farmacologia , Ativador de Plasminogênio Tecidual/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/metabolismo , Ativação Transcricional/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Bovinos , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Linfotoxina-alfa/farmacologia , Proteínas Quinases Ativadas por Mitógeno , NF-kappa B/metabolismo , Ativador de Plasminogênio Tecidual/genética
17.
Invest New Drugs ; 22(1): 17-26, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14707491

RESUMO

The plasminogen activator/plasmin system represents a key component of the proteolytic machinery underlying angiogenesis. In this work, we investigated the effect of Neovastat (AE-941), a naturally occurring multifunctional antiangiogenic agent that is currently in Phase III clinical trials, on tissue and urokinase plasminogen activator activities. We found that in vitro, Neovastat at 100 microg/ml markedly stimulates t-PA-mediated plasmin generation, while it slightly inhibits the generation of plasmin mediated by uPA. The stimulatory effect of Neovastat on t-PA activity was markedly increased by a heat treatment, resulting in a 15-fold increase in the rate of activation of plasminogen. Neovastat did not directly stimulate the activity of t-PA or plasmin towards exogenous substrates, suggesting that its effect requires the presence of plasminogen. Accordingly, kinetic analysis showed that Neovastat increases both the k(cat) of t-PA as well as its affinity for plasminogen by 10-fold. The stimulation of t-PA activity by Neovastat was also correlated with a direct interaction of Neovastat with plasminogen as monitored by the surface plasmon resonance technology. Overall, these results identify Neovastat as a potent stimulator of t-PA-dependent activation of plasminogen, further emphasizing its pleiotropic mechanism of action on several molecular events involved in angiogenesis.


Assuntos
Inibidores da Angiogênese/química , Extratos de Tecidos/química , Ativador de Plasminogênio Tecidual/química , Ativador de Plasminogênio Tipo Uroquinase/química , Catálise , Fibrinolisina/química , Temperatura Alta , Plasminogênio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...