Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Br J Cancer ; 111(1): 139-48, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24874477

RESUMO

BACKGROUND: The elastin-derived peptides (EDPs) exert protumoural activities by potentiating the secretion of matrix metalloproteinases (MMP) and the plasminogen-plasmin activating system. In the present paper, we studied heat-shock protein 90 (Hsp90) involvement in this mechanism. METHODS: HT-1080 fibrosarcoma cell migration and invasion were studied in artificial wound assay and modified Boyden chamber assay, respectively. Heat-shock protein 90 was studied by western blot and immunofluorescence. Matrix metalloproteinase-2 and urokinase plasminogen activator (uPA) were studied by gelatin ± plasminogen zymography and immunofluorescence. Heat-shock protein 90 partners were studied by immunoprecipitation. Messenger RNA expression was studied using real-time PCR. Small interfering RNAs were used to confirm the essential role of Hsp90. RESULTS: We showed that kappa-elastin and VGVAPG elastin hexapeptide stimulated Hsp90, pro-MMP-2 and uPA secretion within 6 h, whereas AGVPGLGVG and GRKRK peptides had no effect. No increase of mRNA level was observed. Heat-shock protein 90-specific inhibitors inhibit EDP-stimulated HT-1080 cell-invasive capacity and restrained EDP-stimulated pro-MMP-2 and uPA secretions. The inhibitory effect was reproduced by using Hsp90-blocking antibody or Hsp90 knockdown by siRNA. Heat-shock protein 90 interacted with and stabilised uPA and pro-MMP-2 in conditioned culture media of HT-1080 fibrosarcoma cells. CONCLUSIONS: Taken together, our results demonstrate that EDPs exert protumoural activities through an Hsp90-dependent mechanism involving pro-MMP-2 and uPA.


Assuntos
Movimento Celular/fisiologia , Elastina/farmacologia , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Proteínas de Choque Térmico HSP90/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
2.
Anal Cell Pathol (Amst) ; 35(4): 267-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22407353

RESUMO

BACKGROUND: Leukemic cell adhesion to proteins of the bone marrow microenvironment provides signals which control morphology, motility and cell survival. We described herein the ability of ethoxyfagaronine (etxfag), a soluble synthetic derivative of fagaronine, to prevent leukemic cell adhesion to fibronectin peptide (FN/V). METHODS: Phosphorylation of fak and pyk2 were evaluated by immunoblotting. Labelled proteins were localized by confocal microscopy. PI 3-kinase activity was evaluated by in vitro kinase assay. RESULTS: Subtoxic concentration of etxfag reduced L1210 cell adhesion to FN/V dependently of ß1 integrin engagement. Etxfag impaired FN-dependent formation of ß1 clustering without modifying ß1 expression at the cell membrane. This was accompanied by a decrease of focal adhesion number, a diminution of fak and pyk2 phosphorylation at Tyr-576, Tyr-861 and Tyr-579, respectively leading to their dissociations from ß1 integrin and inhibition of PI 3-kinase activity. Etxfag also induced a cell retraction accompanied by a redistribution of phosphorylated fak and pyk2 in the perinuclear region and lipid raft relocalization. CONCLUSION: Through its anti-adhesive potential, etxfag, combined with conventional cytotoxic drugs could be potentially designed as a new anti-leukemic drug.


Assuntos
Benzofenantridinas/farmacologia , Fibronectinas/metabolismo , Adesões Focais/efeitos dos fármacos , Integrina beta1/metabolismo , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Integrina beta1/genética , Leucemia L1210/genética , Leucemia L1210/metabolismo , Leucemia L1210/patologia , Leucemia Linfoide/genética , Leucemia Linfoide/metabolismo , Leucemia Linfoide/patologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Microscopia Confocal , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina/metabolismo
3.
Leukemia ; 21(4): 595-603, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17301822

RESUMO

Besides its matrix metalloproteinases inhibitory activity, TIMP-1 exhibits other biological activities such as cell survival and proliferation. The intracellular signalling pathway elicited by TIMP-1 begins to be elucidated. We have shown previously that the caspase-3 and the p38alpha MAP kinase were activated during TIMP-1-induced UT-7 cells erythroid differentiation. In this study, we demonstrated that TIMP-1 differentiating effect can be extended to the IL-3-dependent myeloid murine 32D cell line and human erythroid progenitors derived from cord blood CD34(+) cells. By performing small interfering RNA transfection and using chemical inhibitors, we evidenced that caspase-3 was involved in TIMP-1 differentiating effect. We then identified the MEKK1 kinase as a caspase-3 substrate and demonstrated that the MEKK1/MEK6/p38alpha pathway was activated downstream the caspase-3 in TIMP-1-induced hematopoietic differentiation.


Assuntos
Caspase 3/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , MAP Quinase Quinase 6/metabolismo , MAP Quinase Quinase Quinase 1/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Antígenos CD/fisiologia , Antígenos CD34/fisiologia , Diferenciação Celular , Linhagem Celular , Células-Tronco Hematopoéticas/citologia , Humanos , MAP Quinase Quinase 6/genética , MAP Quinase Quinase Quinase 1/genética , RNA Interferente Pequeno/genética
4.
Leukemia ; 19(3): 373-80, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15674360

RESUMO

The mechanism underlying p210(BCR/ABL) oncoprotein-mediated transformation in chronic myelogenous leukemia (CML) is not fully understood. We hypothesized that p210(BCR/ABL) suppresses expression of genes which may explain at least some of the pathogenetic features of CML. A subtractive cDNA library was created between BCR/ABL-enhanced-green-fluorescent-protein (GFP)-transduced umbilical cord blood (UCB) CD34+ cells and GFP-transduced UCB CD34+ cells to identify genes whose expression is downregulated by p210(BCR/ABL). At least 100 genes were identified. We have confirmed for eight of these genes that expression was suppressed by quantitative real-time-RT-PCR (Q-RT-PCR) of additional p210(BCR/ABL)-transduced CD34+ UCB cells as well as primary early chronic phase (CP) bone marrow (BM) CML CD34+ cells. Imatinib mesylate reversed downregulation of some genes, to approximately normal levels. Several of the genes are implicated in cell adhesion and motility, including L-selectin, intercellular adhesion molecule-1 (ICAM-1), and the chemokine receptor, CCR7, consistent with the known defect in adhesion and migration of CML cells. Compared with GFP UCB or normal (NL) BM CD34+ cells, p210 UCB and CML CD34+ cells migrated poorly towards the CCR7 ligands, CCL19 and CCL21, suggesting a possible role for CCR7 in the abnormal migratory behavior of CML CD34+ cells.


Assuntos
Movimento Celular/fisiologia , Quimiocinas CC/fisiologia , Proteínas de Fusão bcr-abl/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Receptores de Quimiocinas/fisiologia , Adesão Celular/genética , Adesão Celular/fisiologia , Linhagem Celular , Movimento Celular/genética , Quimiocina CCL19 , Quimiocina CCL21 , Quimiocinas CC/genética , Regulação para Baixo , Proteínas de Fusão bcr-abl/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Células K562 , Selectina L/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Ligantes , RNA Mensageiro/genética , Receptores CCR7 , Receptores de Quimiocinas/genética
5.
Leukemia ; 18(4): 727-33, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14961028

RESUMO

Chronic myelogenous leukemia (CML) is a malignancy of the human hematopoietic stem cell (HSC) caused by the p210BCR/ABL oncoprotein. Although alternative splicing of pre-mRNA is a critical determinant of a cell's protein repertoire, it has not been associated with CML pathogenesis. We identified a BCR/ABL-dependent increase in expression of multiple genes involved in pre-mRNA splicing (eg SRPK1, RNA Helicase II/Gu, and hnRNPA2/B1) by subtractive hybridization of cDNA from p210BCR/ABL-eGFP vs eGFP-transduced umbilical cord blood CD34+ cells. beta1-integrin signaling is important to HSC maintenance and proliferation/differentiation, and is abnormal in CML. As an example of how changes in pre-mRNA processing might contribute to CML pathogenesis, we observed alternative splicing of a gene for a beta1-integrin-responsive nonreceptor tyrosine kinase (PYK2), resulting in increased expression of full-length Pyk2 in BCR/ABL-containing cells. Treatment of p210BCR/ABL-positive cells with the Abl-specific tyrosine kinase inhibitor STI571 reverted PYK2 splicing to a configuration more consistent with normal cells, and correlated with decreased expression of BCR/ABL-induced proteins involved in pre-mRNA processing. Whether altered PYK2 splicing contributes to CML pathogenesis remains undetermined; however, we propose that generic changes in pre-mRNA splicing as a result of p210BCR/ABL kinase activity may contribute to CML pathogenesis.


Assuntos
Proteínas de Fusão bcr-abl/fisiologia , Células-Tronco Hematopoéticas/patologia , Proteínas Tirosina Quinases/genética , Splicing de RNA , Antígenos CD34 , Quinase 2 de Adesão Focal , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/etiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide de Fase Crônica/etiologia , Leucemia Mieloide de Fase Crônica/genética , Leucemia Mieloide de Fase Crônica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Precursores de RNA/genética
6.
J Interferon Cytokine Res ; 20(6): 577-87, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10888114

RESUMO

Interferon (IFN) is an effective treatment for chronic myeloid leukemia (CML) in chronic phases, and a number of in vitro antileukemic effects of IFN on CML cells have been reported. The transfer of cytokine genes into tumor cells is reportedly a valuable approach to improve the antitumor activity of cytokines in various models. We first investigated the possibility of transducing CML cells with the retroviral vectors LIalpha2SN and LIgammaSN, encoding the IFN-alpha2 and IFN-gamma genes, respectively, and with the bicistronic vector LIalpha2IrIgammaSN coexpressing the IFN-alpha2 and IFN-gamma genes. We then analyzed the effects of IFN-alpha2 and IFN-gamma produced alone or simultaneously on the proliferation of CML cells. We optimized the transduction efficiency by using the CML-derived K562 cell line. We then introduced IFN genes into CML CD34+ cells. Secretion of IFN-alpha and IFN-gamma was demonstrated in K562 and CML CD34+ cells transduced with the different vectors. The MHC class I antigens were overexpressed in both K562 and CML CD34+ transduced cells. Inhibition of the proliferation of LIalpha2IrIgammaSN-transduced CML cells was greater than with the LIalpha2SN and the LIgammaSN-transduced CML cells. We demonstrate an additive effect of IFN-alpha and IFN-gamma on the inhibition of K562 and CML CD34+ cell proliferation.


Assuntos
Interferon Tipo I/genética , Interferon gama/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Antígenos CD34/metabolismo , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Primers do DNA/genética , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Interferon Tipo I/biossíntese , Interferon Tipo I/farmacologia , Interferon gama/biossíntese , Interferon gama/farmacologia , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Proteínas Recombinantes , Retroviridae/genética , Transdução Genética , Ensaio Tumoral de Célula-Tronco
7.
J Interferon Cytokine Res ; 19(5): 533-41, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10386866

RESUMO

Gene transduction into immature human hematopoietic cells collected from umbilical cord blood, bone marrow, or mobilized peripheral blood cells could be useful for the treatment of genetic and acquired disorders of the hematopoietic system. Immunodeficient mouse models have been used frequently as recipients to assay the growth and differentiation of human hematopoietic stem/progenitor cells. Indeed, high levels of human cell engraftment were first reported in human/murine chimeras using NOD/SCID mice, which now are considered as the standard for these types of experiments. However, NOD/SCID mice have some clear disadvantages (including spontaneous tumor formation) that limit their general use. We have developed a new immunodeficient mouse model by combining recombinase activating gene-2 (RAG2) and common cytokine receptor gamma chain (gamma c) mutations. The RAG2-/-/gamma c- double mutant mice are completely alymphoid (T-, B-, NK-), show no spontaneous tumor formation, and exhibit normal hematopoietic parameters. Interestingly, human cord blood cell engraftment in RAG2-/-/gamma c- mice was greatly enhanced by the exogenous administration of human cytokines interleukin-(IL-3) granulocyte-macrophage colony-stimulating factor, (GM-CSF), and erythropoietin in contrast to the NOD/SCID model. This unique feature of the RAG2-/-/gamma c- mouse model should be particularly well suited for assessing the role of different cytokines in human lymphopoiesis and stem/progenitor cell function in vivo.


Assuntos
Citocinas/farmacologia , Transplante de Células-Tronco Hematopoéticas , Fragmentos de Peptídeos/genética , Receptores de Citocinas/genética , Imunodeficiência Combinada Severa/genética , Animais , Antígenos CD34/sangue , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-3/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação , Proteínas Nucleares , Fenótipo , Receptores de Citocinas/química
8.
Ann Biol Clin (Paris) ; 57(1): 43-50, 1999.
Artigo em Francês | MEDLINE | ID: mdl-9920966

RESUMO

Gene transfer in hematopoietic cells is intended to treat patients with malignant disease and inherited monogenic (hematological, immunological, and metabolic) disorders. Hematopoietic progenitor or stem cells are a favoured target for gene therapy because these cells are easily withdrawn from the patient, expanded and genetically modified ex vivo and then reinjected into the organism. Retroviral vectors allow an efficient transfer of the genes of interest. Transduction of stem cells leads to a stable expression of the transgene for long periods of time. However, we are at the beginning of this new therapeutic application, the technique was being already successful in very few cases. Problems to be solved are mainly in the understanding of the physiology of the hematopoietic stem cell and in the improvement of technical qualities of the vectors for a targeted gene transfer in vivo.


Assuntos
Terapia Genética/métodos , Vírus Defeituosos/genética , Expressão Gênica , Técnicas de Transferência de Genes , Doenças Genéticas Inatas/terapia , Marcadores Genéticos , Vetores Genéticos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Humanos , Neoplasias/terapia , Retroviridae
9.
Gene Ther ; 5(4): 556-62, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9614582

RESUMO

We have developed an efficient and rapid method to analyze transduction in human hematopoietic cells and to select them. We constructed two retroviral vectors using the recombinant humanized S65T green fluorescent protein (rHGFP) gene. Transduced cells appeared with specific green fluorescence on microscopy or fluorescence-activated cell sorting (FACS) analysis. The rHGFP gene was placed under the control of two different retroviral promotors (LTR) in the LGSN vector and in the SF-GFP vector. Amphotropic retroviruses were tested on NIH/3T3 fibroblasts or human hematopoietic (K562, TF-1) cell lines. Then CD34+ cells isolated from cord blood were infected three times after a 48-h prestimulation with IL-3, IL-6, SCF or with IL-3, IL-6, SCF, GM-CSF, Flt3-L and TPO. After 6 days of expansion, a similar number of total CD34(+)-derived cells, CD34+ cells and CFC was obtained in non-transduced and transduced cells, demonstrating the absence of toxicity of the GFP. A transduction up to 46% in total CD34(+)-derived cells and 21% of CD34+ cells was shown by FACS analysis. These results were confirmed by fluorescence of colonies in methyl-cellulose (up to 36% of CFU-GM and up to 25% of BFU-E). The FACS sorting of GFP cells led to 83-100% of GFP-positive colonies after 2 weeks of methyl-cellulose culture. Moreover, a mean gene transfer efficiency of 8% was also demonstrated in longterm culture initiating cells (LTC-IC). This rapid and efficient method represents a substantial improvement to monitor gene transfer and retroviral expression of various vectors in characterized human hematopoietic cells.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Células-Tronco Hematopoéticas , Retroviridae , Transfecção , Células Cultivadas , Citometria de Fluxo , Expressão Gênica , Marcadores Genéticos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Microscopia de Fluorescência
10.
Cancer Gene Ther ; 5(6): 390-400, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9917094

RESUMO

The transfer and expression of cytokine genes into tumor cells is reportedly a valuable approach to improve the antitumor activity of cytokines in various models. Interferon (IFN)-alpha may induce hematological remission in chronic myeloid leukemia (CML) patients, but only a small proportion of patients achieve a sustained, complete cytogenetic remission. We have investigated the possibility of transducing CML cells with the retroviral vector LIalpha2SN, which encodes the IFN-alpha2 gene. We first optimized the transduction efficiency using the CML-derived K562 cell line. A transduction efficiency of 50% and 85% after three and six infections, respectively, was obtained in K562 cells. We then expressed IFN-alpha2 in CML cells by transducing the latter with LIalpha2SN viral particles. The IFN-alpha secretion after three and six infections was 5,400 and 18,000 U/24 hours/10(6) cells for unselected K562 cells and 7,000 and 290 U/24 hours/10(6) cells for CML CD34+ cells at days 4 and 5. Moreover, the major histocompatibility complex class I antigens were overexpressed after infection with LIalpha2SN in both K562 and CML CD34+ cells. The proliferation (in liquid culture) and the cloning efficiency of these CML cells were significantly decreased after LIalpha2SN treatment. By contrast, the proliferation of cord blood CD34+ cells was not affected by transduction with LIalpha2SN. These results demonstrate the transduction efficiency of CML cells and suggest the possibility of CML cell immunotherapy with retroviral gene transfer of different cytokines such as IFN-alpha.


Assuntos
Terapia Genética , Vetores Genéticos , Interferon-alfa/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Retroviridae/genética , Animais , Antígenos CD34/metabolismo , Divisão Celular , Sangue Fetal/imunologia , Citometria de Fluxo , Técnicas de Transferência de Genes , Genes MHC Classe I/efeitos dos fármacos , Humanos , Células K562 , Camundongos , Fatores de Tempo , Transdução Genética
11.
J Inherit Metab Dis ; 20(2): 247-57, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9211197

RESUMO

Congenital erythropoietic porphyria (CEP) is an inherited metabolic disorder characterized by an overproduction and accumulation of porphyrins in bone marrow. This autosomal recessive disease results from a deficiency of uroporphyrinogen III synthase (UROIIIS), the fourth enzyme of the haem biosynthetic pathway. It is phenotypically heterogeneous: patients with mild disease have cutaneous involvement, while more severely affected patients are transfusion dependent. The cloning of UROIIIS cDNA and genomic DNA has allowed the molecular characterization of the genetic defect in a number of families. To date, 22 different mutations have been characterized. Allogeneic bone marrow transplantation is the only curative treatment available for the severe, transfusion-dependent, cases. When bone marrow transplantation cannot be performed owing to the absence of a suitable donor, the autografting of genetically modified cells is an appealing alternative. The best approach to somatic gene therapy in this disease involves the use of recombinant retroviral vectors to transduce cells ex vivo, followed by autologous transplantation of the genetically modified cells. We investigated retroviral transfer in deficient human fibroblasts, immortalized lymphoblasts as well as bone marrow cells, and obtained a complete restoration of the enzymatic activity and full metabolic correction. Using K562 cells, an erythroleukaemic cell line, the expression of the transgene remained stable during 3 months and during erythroid differentiation of the cells. Finally, a 1.6- to 1.9-fold increase in enzyme activity compared to the endogenous level was found in normal CD34+ cells, a population of heterogeneous cells known to contain the progenitor/stem cells for long-term expression. The future availability of a mouse model of the disease will permit ex vivo gene therapy experiments on the entire animal.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Porfiria Eritropoética/terapia , Uroporfirinogênio III Sintetase/genética , Células 3T3 , Animais , Antígenos CD34 , DNA Complementar , Células Precursoras Eritroides , Expressão Gênica , Humanos , Camundongos , Transformação Genética , Células Tumorais Cultivadas
12.
J Med Microbiol ; 36(5): 321-31, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1588582

RESUMO

Detection of an unusual combination of four resistance markers among coagulase-negative staphylococci (CNS) isolated in the same intensive care unit led to the undertaking of an epidemiological assessment. Seventeen CNS isolates from the same unit and 38 epidemiologically unrelated Staphylococcus epidermidis isolates were typed by eight methods, including analysis of immunoblot patterns and hybridisation patterns (HP) obtained with three probes. The probes comprised plasmids carrying the genes encoding 16S rRNA (pBA2), aacA-aphD (pSF815A), and aacA-aphD with part of IS256 (pIP1307). Immunoblot patterns and HP with pIP1307 indicated that 14 of the 17 CNS isolates from the same unit resulted from the spread of an epidemic strain.


Assuntos
Coagulase/metabolismo , Unidades de Terapia Intensiva , Staphylococcus/isolamento & purificação , Proteínas de Bactérias/análise , Métodos Epidemiológicos , Humanos , Immunoblotting , Testes de Sensibilidade Microbiana , Paris , Plasmídeos , Sorotipagem , Staphylococcus/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA