Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Int J Mol Sci ; 22(22)2021 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-34830422

RESUMO

Immortalized cell lines have been used in a wide range of applications in research on immune disorders and cellular metabolic regulation due to the stability and uniformity of their cellular characteristics. At present, the investigation into molecular functions and signaling pathways within bovine cells remains largely limited by the lack of immortalized model cells. Current methods for immortalizing bovine cells are mainly restricted to the ectopic expression of human telomerase reverse transcriptase (hTERT) through transient transfection or virus-mediated delivery, which have defects in efficiency and reliability. In this study, we identified bovine TERT (bTERT) as a novel potent biofactor for immortalizing bovine cells with great advantages over hTERT, and established an efficient and easily manipulated strategy for the immortalization of bovine primary cells. Through the homology-mediated end-joining-based insertion of bTERT at the ROSA26 locus, we successfully generated immortalized bovine fetal fibroblast cell lines with stable characteristics. The observed limitation of this strategy in immortalizing bovine bone marrow-derived macrophages was attributed to the post-translational modification of bTERT, causing inhibited nuclear localization and depressed activity of bTERT in this terminally differentiated cell. In summary, we constructed an innovative method to achieve the high-quality immortalization of bovine primary cells, thereby expanding the prospects for the future application of immortalized bovine model cell lines.


Assuntos
Linhagem Celular Transformada/citologia , Reparo do DNA por Junção de Extremidades/genética , Telomerase/genética , Animais , Bovinos , Linhagem Celular Transformada/enzimologia , Regulação Enzimológica da Expressão Gênica , Humanos , Reparo de DNA por Recombinação/genética
2.
Clin Exp Immunol ; 161(1): 71-80, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20491788

RESUMO

Main features of rheumatoid arthritis (RA), hyperplasia of fibroblast-like synoviocytes (FLS) and joint destruction are caused by inflammatory cytokines produced in chronic autoimmune inflammation. Cell-intrinsic acquisition of tumour-like phenotypes of RA-FLS could also be responsible for the aggressive proliferation and invasion, which are supported by the fact that in some cases RA-FLS has mutations of a tumour suppressor gene TP53. However, the underlying molecular mechanism for TP53 mutations in RA-FLS has not yet been clarified. Recently it has been reported that the non-lymphoid cells in the inflammatory tissues express ectopically the activation-induced cytidine deaminase (AID) gene that induces somatic hypermutations, not only at the immunoglobulin (Ig) gene variable regions in germinal centre B lymphocytes but also at coding regions in TP53. Real-time polymerase chain reaction (PCR) analyses revealed more than half (five of nine) of the RA-FLS lines we established showed the markedly increased expression of AID. AID transcription in RA-FLS was augmented by tumour necrosis factor (TNF)-alpha and even by physiological concentration of beta-oestradiol that could not induce AID transcription in osteoarthritis-FLS. Furthermore, AID-positive RA-FLS presented a higher frequency of somatic mutations in TP53. Cytological and immunohistochemical analyses demonstrated clearly the ectopic expression of AID in the FLS at the RA synovium. These data suggested strongly a novel consequence of RA; the ectopic expression of AID in RA-FLS causes the somatic mutations and dysfunction of TP53, leading to acquisition of tumour-like properties by RA-FLS.


Assuntos
Artrite Reumatoide/patologia , Citidina Desaminase/fisiologia , Genes p53 , Mutação , Membrana Sinovial/enzimologia , Proteína Supressora de Tumor p53/fisiologia , Idoso , Idoso de 80 Anos ou mais , Artrite Reumatoide/enzimologia , Artrite Reumatoide/genética , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/metabolismo , Linhagem Celular Transformada/patologia , Transformação Celular Neoplásica , Sistemas Computacionais , Citidina Desaminase/biossíntese , Indução Enzimática , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperplasia , Masculino , Pessoa de Meia-Idade , Osteoartrite/genética , Osteoartrite/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
3.
Theriogenology ; 71(9): 1417-24, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19303628

RESUMO

Although mammary epithelial cell lines can provide a rapid and reliable indicator of gene expression efficiency of transgenic animals, their short lifespan greatly limits this application. To provide stable and long lifespan cells, goat mammary epithelial cells (GMECs) were transduced with pLNCX2-hTERT by retrovirus-mediated gene transfer. Transduced GMECs were evaluated by reverse transcriptase polymerase chain reaction (RT-PCR), proliferation assays, karyotype analysis, telomerase activity assay, western blotting, soft agar assay, and injection into nude mice. Non-transduced GMECs were used as a control. The hTERT-GMECs had higher telomerase activity and extended proliferative lifespan compared to non-transfected GMECs; even after Passage 50, hTERT-GMECs had a near diploid complement of chromosomes. Furthermore, they did not gain the anchorage-independent growth property and were not associated with a malignant phenotype in vitro or in vivo.


Assuntos
Linhagem Celular Transformada/enzimologia , Cabras , Glândulas Mamárias Animais/citologia , Telomerase/genética , Animais , Divisão Celular , Células Epiteliais , Feminino , Expressão Gênica , Vetores Genéticos , Humanos , Cariotipagem , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telomerase/metabolismo , Transfecção
4.
Oncogene ; 28(2): 279-88, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18836480

RESUMO

The recepteur d'origine nantais (RON) is a receptor tyrosine kinase (RTK) in the scatter factor family, which includes the c-Met receptor. RON exhibits increased expression in a significant number of human breast cancer tissues as well as in many established breast cancer cell lines. Recent studies have indicated that in addition to ligand-dependent signaling events, RON also promotes signals in the absence of its only known ligand, MSP, when expressed in epithelial cells. In this study, we found that when expressed in MCF-10A breast epithelial cells, RON exhibits both MSP-dependent and MSP-independent signaling, which lead to distinct biological outcomes. In the absence of MSP, RON signaling promotes cell survival, increased cell spreading and enhanced migration in response to other growth factors. However, both RON-mediated proliferation and migration require the addition of MSP in MCF-10A cells. Both MSP-dependent and MSP-independent signaling by RON are mediated in part by Src family kinases. These data suggest that RON has two alternative modes of signaling that can contribute to oncogenic behavior in normal breast epithelial cells.


Assuntos
Mama/citologia , Transformação Celular Neoplásica , Células Epiteliais/enzimologia , Fator de Crescimento de Hepatócito/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular/citologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/enzimologia , Linhagem Celular Transformada/citologia , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Forma Celular/efeitos dos fármacos , Forma Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Camundongos , Células NIH 3T3/citologia , Células NIH 3T3/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Proteína Tirosina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/fisiologia
5.
Biotechnol Lett ; 29(11): 1623-30, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17636386

RESUMO

A secretory form of human alpha3-fucosyltransferase IX (sFUT9) was overexpressed in Spodoptera frugiperda (Sf9) insect cells using the stable expression vector pIB/V5-His-TOPO and the signal sequence of human interleukin 2 for efficient secretion. sFUT9 was active and its three potential N-glycosylation sites were occupied. sFUT9 efficiently fucosylated the type II acceptors Galbeta4GlcNAC-R and Fucalpha2Galbeta4GlcNAc-R (R = (CH2)3NHCO(CH2)5-NH-biotin) but not the corresponding sialylated acceptor, and only very poorly the type I (Galbeta3GlcNAc-R) related acceptors. sFUT9 showed a clear preference for glycoproteins containing type II acceptors, with values of 121, 113 and 110 microU/million cell for asialofetuin, erythropoietin and asialoerythropoietin, respectively, values approximately 11-fold higher than those obtained for the small acceptors.


Assuntos
Linhagem Celular Transformada/enzimologia , Fucosiltransferases/biossíntese , Proteínas Recombinantes/biossíntese , Spodoptera , Animais , Células Cultivadas , Vetores Genéticos , Glicosilação , Humanos , Antígenos do Grupo Sanguíneo de Lewis/fisiologia , Spodoptera/enzimologia , Spodoptera/genética , Spodoptera/metabolismo
6.
Leukemia ; 21(3): 421-6, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17252018

RESUMO

Imatinib mesylate is a selective inhibitor of the oncogenic tyrosine kinase, Bcr-Abl, and is widely used as a first-line treatment for chronic myeloid leukaemia (CML). Prolonged monotherapy is frequently associated with patients becoming refractory to imatinib. Therefore, there is considerable interest in small molecule inhibitors which may be used either as replacements or as adjuncts to existing imatinib therapy. For this purpose, it is most likely that drugs which do not share imatinib's mechanism of action will be most valuable. We compared two such compounds with different modes of action, adaphostin and 17-allylamino-17-demethoxygeldanamycin (17-AAG), for their cytotoxic effect and ability to induce the downregulation of cellular proteins in a murine haemopoietic cell line transformed with human p210(Bcr-Abl), and two subclones resistant to imatinib owing to an Abl-kinase domain mutation (E255K) or amplification of the BCR-ABL gene, respectively. We found that, whereas 17-AAG selectively killed Bcr-Abl-positive cells and inhibited proteins dependent on heat-shock protein 90 for their stability (p210(Bcr-Abl) and Akt), adaphostin induced the downregulation of multiple cell-signalling proteins (p210(Bcr-Abl), Akt, Bcr, Abl and STAT5a) and was cytotoxic to both Bcr-Abl-positive and -negative cells. We suggest that both compounds may prove useful in the treatment of CML but caution that undesirable side-effects may result from the inhibition of multiple cell signalling proteins.


Assuntos
Adamantano/análogos & derivados , Antineoplásicos/farmacologia , Benzoquinonas/farmacologia , Hidroquinonas/farmacologia , Lactamas Macrocíclicas/farmacologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Adamantano/efeitos adversos , Adamantano/farmacologia , Animais , Benzamidas , Benzoquinonas/efeitos adversos , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/enzimologia , Células Clonais/efeitos dos fármacos , Células Clonais/enzimologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/biossíntese , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/fisiologia , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Genes abl , Proteínas de Choque Térmico HSP90/fisiologia , Humanos , Peróxido de Hidrogênio/farmacologia , Hidroquinonas/efeitos adversos , Mesilato de Imatinib , Lactamas Macrocíclicas/efeitos adversos , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Mutação de Sentido Incorreto , Estresse Oxidativo/efeitos dos fármacos , Mutação Puntual , Inibidores de Proteínas Quinases/efeitos adversos , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-bcr/biossíntese , Proteínas Proto-Oncogênicas c-bcr/genética , Espécies Reativas de Oxigênio , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/genética , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Transfecção
7.
Biol Reprod ; 76(1): 29-35, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17005940

RESUMO

To study mechanisms governing fetoplacental vascular function, we have established a primary ovine fetoplacental artery endothelial (OFPAE) cell line. These OFPAE cells produce nitric oxide (NO), proliferate, and migrate in response to fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF). To overcome the senescence crisis that this primary OFPAE cell line will eventually enter, we attempted to establish a functional OFPAE cell line with a prolonged life span by transfecting cells with plasmids containing a neomycin resistance gene and a simian virus 40 gene (SV40) expressing large T (T) and small t (t) antigens. The OFPAE cells at passage 8 were transfected. After neomycin selection, the surviving OFPAE (designated SV40 OFPAE) cells were expanded up to passage 80. Up to passage 30, these SV40 OFPAE cells maintained a morphology similar to untransfected OFPAE cells. Expression of T and t antigens in SV40 OFPAE cells was confirmed by immunocytochemistry. These SV40 OFPAE cells exhibited positive uptake of acetylated low-density lipoprotein (Ac-LDL) and positive staining for NO synthase 3 (NOS3) and formed capillary-like tube structures on Matrigel. Up to passages 20-23, these SV40 OFPAE cells proliferated (P < 0.05) and produced (P < 0.05) NO in response to both FGF2 and VEGF. Moreover, this cell proliferation stimulated by FGF2 and VEGF was dose-dependently inhibited (P < 0.05) by PD98059 (a selective mitogen-activated protein kinase 1 and 2 [MAP2K1/2, also termed MEK1/2] inhibitor) or by LY294002 (a selective phosphoinositide 3-kinase [PI3K] inhibitor). These data indicate that SV40 OFPAE cells, at least at passage 23, retain endothelial phenotypes and functions similar to their parental, untransfected OFPAE cells. Thus, a functional OFPAE cell line with an extended life span has been successfully established, potentially providing a valuable cell model for studying fetoplacental endothelial function.


Assuntos
Artérias/citologia , Linhagem Celular Transformada/citologia , Células Endoteliais/citologia , Placenta/irrigação sanguínea , Animais , Artérias/enzimologia , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , Proliferação de Células , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Plasmídeos/genética , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carneiro Doméstico , Vírus 40 dos Símios/genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Biochem J ; 396(1): 23-30, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16478441

RESUMO

CD39/ecto-NTPDase 1 (nucleoside triphosphate diphosphohydrolase 1) is an ecto-nucleotidase that influences P2 receptor activation to regulate vascular and immune cell adhesion and signalling events pivotal in inflammation. Whether CD39 interacts with other membrane or cytoplasmic proteins has not been established to date. Using the yeast two-hybrid system, we note that the N-terminus of CD39 binds to RanBPM (Ran binding protein M; also known as RanBP9), a multi-adaptor scaffolding membrane protein originally characterized as a binding protein for the small GTPase Ran. We confirm formation of complexes between CD39 and RanBPM in transfected mammalian cells by co-immunoprecipitation studies. Endogenous CD39 and RanBPM are also found to be co-expressed and abundant in cell membranes of B-lymphocytes. NTPDase activity of recombinant CD39, but not of N-terminus-deleted-CD39 mutant, is substantially diminished by RanBPM co-expression in COS-7 cells. The conserved SPRY [repeats in splA and RyR (ryanodine receptor)] moiety of RanBPM is insufficient alone for complete physical and functional interactions with CD39. We conclude that CD39 associations with RanBPM have the potential to regulate NTPDase catalytic activity. This intermolecular interaction may have important implications for the regulation of extracellular nucleotide-mediated signalling.


Assuntos
Antígenos CD/fisiologia , Apirase/fisiologia , Linfócitos B/enzimologia , Proteínas Nucleares/fisiologia , Proteína ran de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos CD/química , Apirase/química , Células COS , Linhagem Celular Transformada/enzimologia , Membrana Celular/enzimologia , Chlorocebus aethiops , Proteínas do Citoesqueleto , DNA Complementar/genética , Humanos , Camundongos , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/fisiologia , Relação Estrutura-Atividade , Transfecção , Técnicas do Sistema de Duplo-Híbrido
9.
Mol Cell Biol ; 25(15): 6464-74, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16024784

RESUMO

Murine embryo fibroblasts are readily transformed by the introduction of specific combinations of oncogenes; however, the expression of those same oncogenes in human cells fails to convert such cells to tumorigenicity. Using normal human and murine embryonic fibroblasts, we show that the transformation of human cells requires several additional alterations beyond those required to transform comparable murine cells. The introduction of the c-Myc and H-RAS oncogenes in the setting of loss of p53 function efficiently transforms murine embryo fibroblasts but fails to transform human cells constitutively expressing hTERT, the catalytic subunit of telomerase. In contrast, transformation of multiple strains of human fibroblasts requires the constitutive expression of c-Myc, H-RAS, and hTERT, together with loss of function of the p53, RB, and PTEN tumor suppressor genes. These manipulations permit the development of transformed human fibroblasts with genetic alterations similar to those found associated with human cancers and define specific differences in the susceptibility of human and murine fibroblasts to experimental transformation.


Assuntos
Linhagem Celular Transformada/patologia , Fibroblastos/patologia , Proteínas Oncogênicas Virais/fisiologia , Animais , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/virologia , Transformação Celular Neoplásica/patologia , Inibidor p16 de Quinase Dependente de Ciclina , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/virologia , Humanos , Camundongos , Proteínas Oncogênicas Virais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/fisiologia , Telomerase/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/metabolismo
10.
Hum Mutat ; 25(3): 239-47, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15714522

RESUMO

The cblE type of homocystinuria is a rare autosomal recessive disorder caused by impaired reductive activation of methionine synthase. Although earlier biochemical studies proposed that the methionine synthase enzyme might be activated by two different reducing systems, mutations were reported in only the methionine synthase reductase gene (MTRR) in cblE patients. The pathogenicity of MTRR mutations, however, has not yet been tested functionally. We report on nine patients of European origin affected by the cblE type of homocystinuria. They presented between 2 weeks and 3 years of age (median age 4 weeks) with anemia, which was macrocytic in only three patients, and with neurological involvement in all but two cases. Bone marrow examination performed in seven patients showed megaloblastic changes in all but one of them. All patients exhibited moderate to severe hyperhomocysteinemia (median plasma total homocysteine [Hcy] 92 mumol/L, range 44-169), while clearly reduced methionine was observed only in four cases. Pathogenic mutations were identified in both parental alleles of the MTRR gene in all patients. Five known (c.903+469T>C, c.1361C>T, c.1459G>A, c.1557-4_1557+3del7, and c.1622_1623dupTA) and three novel mutations (c.7A>T, c.1573C>T, and c.1953-6_1953-2del5) were detected. Importantly, transfection of fibroblasts of cblE patients with a wild-type MTRR minigene expression construct resulted in a significant approximately four-fold increase of methionine synthesis, indicating correction of the enzyme defect. Our study shows a link between a milder predominantly hematological presentation and homozygosity for the c.1361C>T mutation, but no other obvious genotype-phenotype correlation. The identification of mutations in the MTRR gene, together with restoration of methionine synthesis following MTRR minigene expression in cblE cells confirms that this disease is caused by defects in the MTRR gene.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/deficiência , Ferredoxina-NADP Redutase/deficiência , Terapia Genética , Homocistinúria/genética , Substituição de Aminoácidos , Betaína/uso terapêutico , Encéfalo/patologia , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/patologia , Códon sem Sentido , Análise Mutacional de DNA , Ferredoxina-NADP Redutase/genética , Fibroblastos/enzimologia , Fibroblastos/patologia , Ácido Fólico/uso terapêutico , Genes Sintéticos , Teste de Complementação Genética , Haplótipos/genética , Homocisteína/sangue , Homocistinúria/sangue , Homocistinúria/classificação , Homocistinúria/tratamento farmacológico , Homocistinúria/enzimologia , Homocistinúria/patologia , Homocistinúria/terapia , Humanos , Hidroxocobalamina/uso terapêutico , Mutação de Sentido Incorreto , Mutação Puntual , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Transfecção , População Branca/genética
11.
Expert Opin Biol Ther ; 4(9): 1423-34, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15335310

RESUMO

Chronic granulomatous disease (CGD) is a congenital immune deficiency that is a promising therapeutic target for gene replacement into haematopoietic stem cells (HSCs). CGD results from mutations in any one of four genes encoding subunits of the superoxide-generating NADPH oxidase of phagocytes. Life-threatening, recurrent bacterial and fungal infections, as well as inflammatory granulomas, are the hallmarks of the disease. NADPH oxidase activity can be reconstituted by retroviral- or lentiviral-mediated gene transfer to human CGD marrow in vitro and in xenograft transplant models. Gene transfer studies in knockout mouse models that resemble the human disease suggest that correction of oxidase activity in a minority of phagocytes will be of clinical benefit. Phase I clinical studies in unconditioned CGD patients showed transient expression of small numbers of gene-corrected neutrophils. Areas of research at present include efforts to enhance gene transfer rates into repopulating HSCs using vectors that transduce quiescent cells, and to increase the engraftment of genetically corrected HSCs using non-myeloablative conditioning and drug resistance genes for selection.


Assuntos
Terapia Genética , Doença Granulomatosa Crônica/terapia , Animais , Células da Medula Óssea/enzimologia , Transplante de Medula Óssea , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/transplante , Células Cultivadas/enzimologia , Células Cultivadas/transplante , Ensaios Clínicos Fase I como Assunto , Suscetibilidade a Doenças , Mecanismo Genético de Compensação de Dose , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Doença Granulomatosa Crônica/complicações , Doença Granulomatosa Crônica/genética , Doença Granulomatosa Crônica/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Infecções/etiologia , Inflamação/etiologia , Camundongos , Camundongos Knockout , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Fagócitos/enzimologia , Fagocitose , Recidiva
12.
Cytogenet Genome Res ; 104(1-4): 87-94, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15162019

RESUMO

Ectopic expression of telomerase results in an immortal phenotype in various types of normal cells, including primary human fibroblasts. In addition to its role in telomere lengthening, telomerase has now been found to have various functions, including the control of DNA repair, chromatin modification, and the control of expression of genes involved in cell cycle regulation. The investigations on the long-term effects of telomerase expression in normal human fibroblast highlighted that these cells show low frequencies of chromosomal aberrations. In this paper, we describe the karyotypic stability of human fibroblasts immortalized by expression of hTERT. The ectopic overexpression of telomerase is associated with unusual spontaneous as well as radiation-induced chromosome stability. In addition, we found that irradiation did not enhance plasmid integration in cells expressing hTERT, as has been reported for other cell types. Long-term studies illustrated that human fibroblasts immortalized by telomerase show an unusual stability for chromosomes and for plasmid integration sites, both with and without exposure to ionizing radiation. These results confirm a role for telomerase in genome stabilisation by a telomere-independent mechanism and point to the possibility for utilizing hTERT-immortalized normal human cells for the study of gene targeting.


Assuntos
Cromossomos Humanos/efeitos da radiação , Fibroblastos/efeitos da radiação , Telomerase/fisiologia , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/efeitos da radiação , Linhagem Celular Transformada/ultraestrutura , Aberrações Cromossômicas , Cromossomos Humanos/metabolismo , Células Clonais/enzimologia , Células Clonais/efeitos da radiação , Células Clonais/ultraestrutura , Proteínas de Ligação a DNA , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Marcação de Genes , Humanos , Cariotipagem , Plasmídeos/genética , Tolerância a Radiação , Proteínas Recombinantes de Fusão/fisiologia , Telomerase/genética , Telômero/ultraestrutura , Transfecção , Neoplasias da Bexiga Urinária/patologia
13.
Neuroreport ; 15(2): 245-9, 2004 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15076745

RESUMO

It is necessary to expand human neural progenitor cells in vitro to obtain large numbers for research purposes and cell transplantation. A potential obstacle to in vitro expansion, however, is that neural progenitor cells have a limited replication life-span and gradually lose their differentiation potential. We report here that ectopic expression of the catalytic subunit of human telomerase (hTERT) gene in neural progenitor cells could induce telomerase activity, stabilize telomeres and extend their replicative life-spans. The telomerase-immortalized cells (hNPC-TERT) maintained the normal diploid karyotype, expressed the markers of human neural progenitor cells and meanwhile held the differentiation potential in vitro for up to 120 population doublings. This study provides a new approach for obtaining unlimited quantities of normal phenotypic and homogeneous human neural progenitor cells in vitro.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular Transformada/enzimologia , Neurônios/enzimologia , Células-Tronco/enzimologia , Telomerase/genética , Transdução Genética/métodos , Animais , Biomarcadores , Domínio Catalítico/genética , Diferenciação Celular/genética , Divisão Celular/genética , Linhagem Celular Transformada/citologia , Senescência Celular/genética , Vetores Genéticos/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Cariotipagem , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/citologia , Fenótipo , Células-Tronco/citologia , Telômero/genética , Transgenes/genética
14.
Biochemistry (Mosc) ; 69(3): 306-10, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15061698

RESUMO

We studied comparative expression and activity of cytochrome P450 family 1 (CYP1) isoforms in rat embryo cells, both primary and immortalized by Rausher leukemia virus (RLV). In RLV-infected embryonal cells compared with the initial ones the expression levels of CYP1A1 and 1B1 mRNAs and benzo[a]pyrene (BP) hydroxylase activity were higher, regardless of their treatment with the CYP1 inducer 2,3,7,8-tetrachlorodibenzo-p-dioxin. The sensitivity to BP and 7,12-dimethylbenzo[a]anthracene was higher in the cells immortalized with RLV. The expression level of mRNAs of induction-mediating proteins aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator was the same in both cell cultures tested. Higher sensitivity of cells immortalized with RLV compared with the initial embryo cells to transforming effect of BP, which was described previously, is possibly associated with elevated expression of CYP1 isoforms.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Linhagem Celular Transformada/enzimologia , Transformação Celular Viral , Citocromo P-450 CYP1A1/biossíntese , Vírus Rauscher , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Translocador Nuclear Receptor Aril Hidrocarboneto , Linhagem Celular Transformada/virologia , Transformação Celular Viral/efeitos dos fármacos , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1 , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Dibenzodioxinas Policloradas/farmacologia , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos F344 , Receptores de Hidrocarboneto Arílico/biossíntese , Receptores de Hidrocarboneto Arílico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teratogênicos/farmacologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
15.
Exp Cell Res ; 294(2): 559-70, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15023542

RESUMO

Human Neural Stem Cells (hNSCs) are excellent candidates for in vitro and in vivo molecular, cellular, and developmental research, and also for ex-vivo gene transfer and cell therapy in the nervous system. However, hNSCs are mortal somatic cells, and thus invariably enter an irreversible growth arrest after a finite number of cell divisions in culture. It has been proposed that this is due to telomere shortening. Here, we show that long-term cultured (up to 4 years) v-myc perpetuated hNSC lines do preserve short but stable and homogeneous telomeres (TRF and Q-FISH determinations). hNSC lines (but not strains) express high levels of telomerase activity, which is activated by v-myc, as demonstrated here. Telomerase activity is not constitutive, becoming non-detectable after differentiation (in parallel to v-myc down-regulation). hNSC lines also maintain a stable cell cycle length, mitotic potential, differentiation and neuron generation capacity, and do not express senescence-associated beta-galactosidase over years, as studied here. These data, collectively, help to explain the immortal nature of v-myc-perpetuated hNSC lines, and to establish them as excellent research tools for basic and applied neurobiological and translational studies.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular Transformada/enzimologia , Senescência Celular/genética , Neurônios/enzimologia , Células-Tronco Pluripotentes/enzimologia , Telomerase/metabolismo , Telômero/enzimologia , Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular Transformada/citologia , Sobrevivência Celular/genética , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes myc/genética , Humanos , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Estabilidade de RNA/genética , RNA Mensageiro/metabolismo , Telomerase/genética , Telômero/genética
16.
Toxicol Ind Health ; 20(1-5): 57-68, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15807409

RESUMO

An in vitro study was conducted to determine the effects of variable concentrations of trace metals on human cultured mammary cells. Monolayers of human mortal (MCF-12A) and immortal (MDA-MB231) mammary epithelial cells were incubated in the absence or presence of increasing concentrations of arsenic (As), mercury (Hg) and copper (Cu) for 24-h, 72-h, 4-d, and 7-d. The MTT assay was used to assess viability for all time periods and cell proliferation was monitored for 4-d and 7-d studies. Monolayers were also labeled with rhodamine-110 (R-6501), Sytox green, and Celltiter blue fluorescent dyes as indicators for intracellular esterase activity, nucleic acid staining, and cell reduction/viability, respectively. Total incubation time with chemical plus dyes was 24 h. For 24-h and 72-h studies, cells were seeded in 96-well plates, after which confluent monolayers were exposed to increasing concentrations of chemicals. For 4-d and 7-d studies, cells were seeded in 12-well plates at 1/3 confluent density (day 0) and exposed to increasing concentrations of metals on day 1. All cells were counted on days 4 and 7. In addition, test medium was removed from select groups of cultures on day 4, replaced with fresh medium in the absence of chemical (recovery studies), and assays were performed on day 7 as above. The data suggest that there is a consistent protective and/or stimulating effect of metals at the lowest concentrations in MCF-12A cells that is not observed in immortal MDA-MB231 cells. In fact, cell viability of MCF-12A cells is stimulated by otherwise equivalent inhibitory concentrations of As, Cu, and Hg on MDA-MB231 cells at 24-h. Whereas As and Hg suppress proliferation and viability in both cell lines after 4-d and 7-d of exposure, Cu enhances cell proliferation and viability of MCF-12A cells. MDA-MB231, however, recover better after 4-days of toxic insult. In addition, nutritional manipulation of media between the cell lines, or pretreatment with penicillamine, did not alter the hormesis effect displayed by MCF-12A. Growth of these cells however was not maintained in the alternative medium. The study demonstrates that a hormesis effect from trace metals is detectable in cultured mammary cells; fluorescent indicators, however, are not as sensitive as cell proliferation or MTT in recognizing the subtle responses. Also, sensitivity of mammary cells to lower concentrations of Cu, a biologically important trace metal, may play an important role in controlling cellular processes and proliferation. The ability to detect this in vitro phenomenon implies that similar processes, occurring in vivo, may be responsible for the development, induction, or enhancement of human cancers.


Assuntos
Linhagem Celular Transformada/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Oligoelementos/toxicidade , Arsênio/toxicidade , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/patologia , Sobrevivência Celular/efeitos dos fármacos , Cobre/toxicidade , Relação Dose-Resposta a Droga , Esterases/metabolismo , Feminino , Formazans/metabolismo , Humanos , Mercúrio/toxicidade , Sais de Tetrazólio/metabolismo
17.
Int J Oncol ; 23(2): 477-82, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12851698

RESUMO

Imidazolium trans-imidazole dimethyl sulfoxide tetrachlororuthenate (NAMI-A) is a new compound active against lung metastasis of solid metastasizing tumours. While its in vivo effect has been studied, the molecular insights that underlie its action are largely unknown. Among the possible pathways responsible for malignant transformation, PKC arose as one of the most promising targets for new antineoplastic drugs. We demonstrated the capability of NAMI-A of inhibiting PMA induced-PKC activity in ECV304 in a dose-dependent fashion. Furthermore, NAMI-A through modulation of PKC activity has been proved capable of reducing the phorbol ester induced expression of ornithine decarboxilase (ODC) gene and to abrogate the activation of the Raf/MEK/ERK pathway. Taken together these results suggest that many of the in vivo outcomes of NAMI-A treatment may be the result of a direct action on PKC.


Assuntos
Linhagem Celular Transformada/enzimologia , Dimetil Sulfóxido/análogos & derivados , Dimetil Sulfóxido/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Ornitina Descarboxilase/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/antagonistas & inibidores , Western Blotting , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ornitina Descarboxilase/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Rutênio , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
18.
Cancer Res ; 63(12): 3247-56, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12810655

RESUMO

Demonstration of the existence of G-quadruplex structures in telomeres of Stylonychia macronuclei and in the promoter of c-myc in human cells has validated these secondary DNA structures as potential targets for drug design. The next important issue is the selectivity of G-quadruplex-interactive agents for the different types of G-quadruplex structures. In this study, we have taken an important step in associating specific biological effects of these drugs with selective interaction with either intermolecular or intramolecular G-quadruplex structures formed in telomeres. Telomestatin is a natural product isolated from Streptomyces anulatus 3533-SV4 and has been shown to be a very potent telomerase inhibitor through its G-quadruplex interaction. We have demonstrated that telomestatin interacts preferentially with intramolecular versus intermolecular G-quadruplex structures and also has a 70-fold selectivity for intramolecular G-quadruplex structures over duplex DNA. Telomestatin is able to stabilize G-quadruplex structures that are formed from duplex human telomeric DNA as well as from single-stranded DNA. Importantly, telomestatin stabilizes these G-quadruplex structures in the absence of monovalent cations, which is a unique characteristic among G-quadruplex-interactive compounds. At noncytotoxic concentrations, telomestatin suppresses the proliferation of telomerase-positive cells within several weeks. In contrast, TMPyP4, a compound that preferentially facilitates the formation of intermolecular G-quadruplex structures, suppresses the proliferation of alternative lengthening of telomeres (ALT)-positive cells as well as telomerase-positive cells. We have also demonstrated that TMPyP4 induces anaphase bridges in sea urchin embryos, whereas telomestatin did not have this effect, leading us to conclude that the selectivity of telomestatin for intramolecular G-quadruplex structures and TMPyP4 for intermolecular G-quadruplex structures is important in mediating different biological effects: stabilization of intramolecular G-quadruplex structures produces telomerase inhibition and accelerated telomere shortening, whereas facilitation of the formation of intermolecular G-quadruplex structures induces the formation of anaphase bridges.


Assuntos
Inibidores Enzimáticos/farmacologia , Conformação de Ácido Nucleico , Oxazóis/farmacologia , Porfirinas/farmacologia , Telomerase/antagonistas & inibidores , Telômero/efeitos dos fármacos , Anáfase/efeitos dos fármacos , Animais , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , DNA/efeitos dos fármacos , DNA/ultraestrutura , Humanos , Substâncias Macromoleculares , Potássio/farmacologia , Ouriços-do-Mar/embriologia , Endonucleases Específicas para DNA e RNA de Cadeia Simples/metabolismo , Sódio/farmacologia , Especificidade por Substrato , Telômero/ultraestrutura
19.
Mol Cell Biol ; 23(6): 2096-108, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12612081

RESUMO

The human protein tyrosine phosphatase TCPTP exists as two forms: an endoplasmic reticulum-targeted 48-kDa form (TC48) and a nuclear 45-kDa form (TC45). Although targeted to the nucleus, TC45 can exit in response to specific stimuli to dephosphorylate cytoplasmic substrates. In this study, we investigated the downregulation of insulin receptor (IR) signaling by TCPTP. In response to insulin stimulation, the TC48-D182A and TC45-D182A "substrate-trapping" mutants formed stable complexes with the endogenous tyrosine-phosphorylated IR beta-subunit in 293 cells. Moreover, in response to insulin stimulation, the TC45-D182A mutant accumulated in the cytoplasm of cells overexpressing the IR and in part colocalized with the IR beta-subunit at the cell periphery. These results indicate that the IR may serve as a cellular substrate for both TC48 and TC45. In immortalized TCPTP(-/-) murine embryo fibroblasts, insulin-induced IR beta-subunit tyrosine phosphorylation and protein kinase PKB/Akt activation were enhanced relative to the values in TCPTP(+/+) cells. Importantly, the expression of TC45 or TC48 to physiological levels suppressed the enhanced insulin-induced signaling in TCPTP(-/-) cells. These results indicate that the differentially localized variants of TCPTP may dephosphorylate the IR and downregulate insulin-induced signaling in vivo.


Assuntos
Insulina/farmacologia , Isoenzimas/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Fosfatases/fisiologia , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células CHO/efeitos dos fármacos , Células CHO/enzimologia , Bovinos , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/enzimologia , Núcleo Celular/enzimologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Cricetinae , Cricetulus , Citoplasma/enzimologia , Retículo Endoplasmático/enzimologia , Ativação Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Teste de Complementação Genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Sistema de Sinalização das MAP Quinases , Substâncias Macromoleculares , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteína Tirosina Fosfatase não Receptora Tipo 2 , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor de Insulina/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo
20.
Oncogene ; 22(7): 1024-34, 2003 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-12592389

RESUMO

Increased expression of mac25/insulin-like growth factor binding-protein related protein-1 (IGFBP-rP1) in human breast and prostate epithelial cell lines results in the suppression of tumor growth. CDNA expression array analysis revealed increased manganese superoxide dismutase (SOD-2) expression in the mac25/IGFBP-rP1-transfected M12 human prostate cancer cell line compared to M12 control cells. SOD-2 has been postulated to be a tumor suppressor. SOD-2 was also increased in LNCaP cells stably transfected with mac25/IGFBP-rP1, but not in mac25/IGFBP-rP1-transfected PC-3 cells. Mac25 LNCaP cells had a marked decrease in tumor growth in nude mice compared to controls, but there was no difference in tumor growth in mac25 PC-3 cells compared to control. Phosphorylated Erk and Akt were increased in the M12 and LNCaP transfected mac25/IGFBP-rP1 cells but not PC-3 mac25. Inhibition of PI-3 kinase results in a marked decrease in viability of the M12-mac25 cells compared to M12 controls. Cells treated with H(2)O(2) result in an increase in phospho-ERK. Transfection of SOD-2 in M12 cells markedly decreased tumor growth, apoptosis, G1 delay in the cell cycle, and expression of senescence associated beta-galactosidase. These results suggest that one of the downstream mediators of the senescence-associated tumor suppression effect of mac25/IGFBP-rP1 is SOD-2.


Assuntos
Adenocarcinoma/patologia , Proteínas de Transporte/fisiologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Proteínas de Neoplasias/fisiologia , Neoplasias da Próstata/patologia , Transdução de Sinais/fisiologia , Superóxido Dismutase/fisiologia , Adenocarcinoma/enzimologia , Animais , Apoptose/fisiologia , Linhagem Celular Transformada/enzimologia , Linhagem Celular Transformada/transplante , Senescência Celular , Cromonas/farmacologia , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fase G1/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Regiões Promotoras Genéticas , Neoplasias da Próstata/enzimologia , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/fisiologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Transfecção , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/transplante , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...