Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.693
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Eur Arch Psychiatry Clin Neurosci ; 270(1): 119-126, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30560291

RESUMO

Repetitive transcranial magnetic stimulation (rTMS) is a neuromodulation technique that stimulates cortical regions via time-varying electromagnetic fields; in several countries this technique has been approved as a treatment for major depressive disorder. One empirically established target in antidepressant pharmacotherapy is the flavin-containing monoamine oxidoreductase (MAO). The function of MAO enzymes is based on oxidation processes that may be sensitive towards strong electromagnetic fields. Therefore, we hypothesized that rTMS-induced electromagnetic fields impact the activity of this enzyme. Using crude synaptosomal cell preparations from human SH-SY5Y neuroblastoma cells and rat cortex as well as viable cells, we assessed the effects of rTMS on MAO-A and -B activity in a well-controlled in vitro set up. In short, samples were stimulated at maximal intensity with an equal number of total stimuli at frequencies of 5, 20, and 100 Hz. Sham stimulation was performed in parallel. Treatment at frequencies of 5 and 20 Hz significantly decreased mainly MAO-B activity in all tissue preparations and species, whereas 100 Hz stimulation remained without effect on any MAO activity. Our results support the hypothesis, that rTMS-induced electromagnetic fields affect MAO activity and provide further evidence for intracellular effects possibly contributing to therapeutic effects of this neuromodulatory method. On a cautionary note, however, our findings are solely based on in vitro evidence.


Assuntos
Córtex Cerebral/enzimologia , Monoaminoxidase/metabolismo , Sinaptossomos/enzimologia , Estimulação Magnética Transcraniana , Células Tumorais Cultivadas/enzimologia , Animais , Linhagem Celular Tumoral , Humanos , Neuroblastoma/enzimologia , Ratos
2.
Croat Med J ; 55(3): 206-17, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24891279

RESUMO

AIM: To use the antioxidant compounds (sodium selenite, selenomethionine, D-pantethine) for modulation of cytotoxic effect of doxorubicin and cisplatin toward wild type and drug-resistant mutants of several human tumor cells. Similar treatments were applied in vivo toward adult male Wistar rats. METHODS: Human tumor cells of different lines (HCT-116, Jurkat and HL-60) with various mechanisms of drug-resistance were treated with doxorubicin or cisplatin, alone or in combination with sodium selenite, selenomethionine, or D-pantethine. Cell viability, induction of apoptosis, and production of O2- radicals were measured. Activity of redox potential modulating enzymes was measured in the liver and blood plasma of adult male Wistar rats subjected to similar treatments. RESULTS: All antioxidants used in physiologically harmless concentration inhibited cytotoxic action of doxorubicin toward tumor cells sensitive to chemotherapy treatment by 15%-30%, and slightly enhanced cytotoxic effect of this medicine toward drug-resistant malignant cells. At the same time, there was no significant effect of these antioxidants on cisplatin action. Such effects were accompanied by a complete inhibition of production of superoxide radicals induced by doxorubicin. The results of in vivo study in adult male Wistar rats were in agreement with the results of in vitro study of human tumor cells. CONCLUSION: Protective effect of specific antioxidant agents during cytotoxic action of doxorubicin was demonstrated in vitro in drug-sensitive human tumor cells and in adult male Wistar rats, while there was no protective effect in drug-resistant sub-lines of these tumor cells during action of doxorubicin and cisplatin.


Assuntos
Antineoplásicos/toxicidade , Antioxidantes/farmacologia , Cisplatino/toxicidade , Doxorrubicina/toxicidade , Panteteína/análogos & derivados , Selenometionina/farmacologia , Selenito de Sódio/farmacologia , Animais , Apoptose/efeitos dos fármacos , Sinergismo Farmacológico , Células HCT116 , Células HL-60 , Humanos , Células Jurkat , Masculino , Oxirredução , Oxirredutases/metabolismo , Panteteína/farmacologia , Ratos , Ratos Wistar , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
3.
Br J Haematol ; 155(2): 198-208, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21848891

RESUMO

Recent findings have indicated that tyrosine kinase inhibitors (TKIs) targeting the ERBB receptor family display anti-leukaemic effects, despite the lack of receptor expression on human leukaemic cells. The occurrence of activating mutations in the gene encoding FMS-like tyrosine kinase 3 (FLT3) in patients with acute myeloid leukaemia (AML) has rendered inhibition of this receptor a promising therapeutic target. Due to possibility of cross-reactivity, we investigated the effect of the irreversible pan-ERBB inhibitor canertinib (CI-1033) on leukaemic cells expressing FLT3. The drug had anti-proliferative and apoptotic effects on primary AML cells and human leukaemic cell lines expressing mutated FLT3. In several AML patient samples, a blast cell population expressing FLT3-internal tandem duplication (ITD) was eradicated by canertinib. Canertinib inhibited receptor autophosphorylation and kinase activity of both mutated and FLT3 ligand stimulated wildtype FLT3, leading to inhibition of the PI3-kinase and MAP kinase pathways. Apoptotic induction was dependent on pro-apoptotic BH3-only protein BCL2L11/BIM because siRNA silencing attenuated apoptosis. Moreover, the drug induced regression of cells expressing FLT3-ITD in a murine in vivo-transplantation model at previously described tolerated doses. These results indicate that canertinib, as an irreversible TKI, could constitute a novel treatment regimen in patients with mutated or overexpressed FLT3.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Morfolinas/uso terapêutico , Proteínas Oncogênicas v-erbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Humanos , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos DBA , Morfolinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/enzimologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sequências de Repetição em Tandem , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia , Tirosina Quinase 3 Semelhante a fms/genética
4.
Eur J Haematol ; 84(3): 239-51, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19922462

RESUMO

OBJECTIVES: Angiogenesis seems important for both leukemogenesis and chemosensitivity in acute myelogenous leukemia (AML). Angiogenesis is regulated by the balance between pro- and antiangiogenic cytokines, which also indicates an important role of matrix metalloproteases (MMPs) and their natural inhibitors, tissue inhibitors of metalloproteases (TIMPs). We investigated the constitutive release of MMPs and TIMPs for a large group of consecutive AML patients. METHODS: AML cells were cultured in vitro either alone or together with microvascular endothelial cells, and levels of MMPs and TIMPs were determined in culture supernatants. RESULTS: AML cells showed constitutive release of several MMPs and TIMPs. For all patients, detectable MMP-10 release was observed, and most patients showed detectable release of at least one additional MMP, usually MMP-9 or MMP-2. A significant correlation was found between MMP-9 and TIMP-1 release and the release of several CCL and CXCL chemokines. MMP-9 release was higher for AML cells with monocytic differentiation corresponding to the FAB-subtype M4/M5 AML; it was mainly released in its inactive form, but endogenously active MMP-9 could be detected even in the presence of the constitutively released TIMP-1/2. Endothelial cells released relatively high levels of MMP-10, and these levels were further increased by coculture with AML cells. Patients achieving complete hematological remission after only one induction cycle showed relatively low constitutive MMP-2 release. CONCLUSION: We conclude that primary human AML cells show constitutive release of both MMPs and TIMPs, and this release may be important for leukemogenesis and possibly also for chemosensitivity.


Assuntos
Leucemia Mieloide/patologia , Metaloproteinases da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Angiopoietina-2/farmacologia , Antraciclinas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácidos Borônicos/farmacologia , Bortezomib , Células Cultivadas/citologia , Quimiocinas/metabolismo , Técnicas de Cocultura , Meios de Cultura Livres de Soro/farmacologia , Citarabina/administração & dosagem , Diterpenos/farmacologia , Células Endoteliais/citologia , Feminino , Humanos , Imidazóis/farmacologia , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/enzimologia , Masculino , Inibidores de Metaloproteinases de Matriz , Pessoa de Meia-Idade , NF-kappa B/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Quinoxalinas/farmacologia , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/fisiologia , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/metabolismo
5.
Biochim Biophys Acta ; 1783(12): 2294-300, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18775751

RESUMO

The present investigation was undertaken to measure the relative abilities of pro-death versus pro-survival proteases in degrading each other and to determine how this might influence cellular susceptibility to death. For this, we first carried out in vitro experiments in which recombinant pro-death proteases (caspase-3 or cathepsin D) were incubated with the pro-survival protease (cathepsin L) in their respective optimal conditions and determined the effects of these reactions on enzyme integrity and activity. The results indicated that cathepsin L was able to degrade cathepsin D, which in turn cleaves caspase-3, however the later enzyme was unable to degrade any of the cathepsins. The consequences of this proteolytic sequence on cellular ability to undergo apoptosis or other types of cell death were studied in cells subjected to treatment with a specific inhibitor of cathepsin L or the corresponding siRNA. Both treatments resulted in suppression of cellular proliferation and the induction of a cell death with no detectable caspase-3 activation or DNA fragmentation, however, it was associated with increased accumulation of cathepsin D, cellular vaculolization, expression of the mannose-6-phosphate receptor, and the autophagy marker LC3-II, all of which are believed to be associated with autophagy. Genetic manipulations leading either to the gain or loss of cathepsin D expression implicated this enzyme as a key player in the switch from apoptosis to autophagy. Overall, these findings suggest that a hierarchy between pro-survival and pro-death proteases may have important consequences on cell fate.


Assuntos
Apoptose/fisiologia , Autofagia , Caspase 3/fisiologia , Catepsina D/fisiologia , Catepsinas/fisiologia , Cisteína Endopeptidases/fisiologia , Western Blotting , Catepsina L , Sobrevivência Celular , Humanos , Microscopia de Fluorescência , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/patologia
6.
J Cell Biol ; 108(5): 1987-95, 1989 May.
Artigo em Inglês | MEDLINE | ID: mdl-2523891

RESUMO

Human HT-1080 fibrosarcoma cells produce urokinase-type plasminogen activator (u-PA) and type 1 plasminogen activator inhibitor (PAI-1). We found that after incubation of monolayer cultures with purified native human plasminogen in serum-containing medium, bound plasmin activity could be eluted from the cells with tranexamic acid, an analogue of lysine. The bound plasmin was the result of plasminogen activation on the cell surface; plasmin activity was not taken up onto cells after deliberate addition of plasmin to the serum-containing medium. The cell surface plasmin formation was inhibited by an anticatalytic monoclonal antibody to u-PA, indicating that this enzyme was responsible for the activation. Preincubation of the cells with diisopropyl fluorophosphate-inhibited u-PA led to a decrease in surface-bound plasmin, indicating that a large part, if not all, of the cell surface plasminogen activation was catalyzed by surface-bound u-PA. In the absence of plasminogen, most of the cell surface u-PA was present in its single-chain proenzyme form, while addition of plasminogen led to formation of cell-bound two-chain u-PA. The latter reaction was catalyzed by cell-bound plasmin. Cell-bound u-PA was accessible to inhibition by endogenous PAI-1 and by added PAI-2, while the cell-bound plasmin was inaccessible to serum inhibitors, but accessible to added aprotinin and an anticatalytic monoclonal antibody. A model for cell surface plasminogen activation is proposed in which plasminogen binding to cells from serum medium is followed by plasminogen activation by trace amounts of bound active u-PA, to form bound plasmin, which in turn serves to produce more active u-PA from bound pro-u-PA. This exponential process is subject to regulation by endogenous PAI-1 and limited to the pericellular space.


Assuntos
Fibrossarcoma/enzimologia , Peptídeo Hidrolases/metabolismo , Ativadores de Plasminogênio/metabolismo , Plasminogênio/metabolismo , Células Tumorais Cultivadas/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Linhagem Celular , Membrana Celular/enzimologia , Meios de Cultura , Ativação Enzimática , Fibrinolisina/metabolismo , Humanos , Cinética , Peso Molecular , Ligação Proteica
7.
J Cell Biol ; 131(3): 761-73, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7593195

RESUMO

The NBT-II rat carcinoma cell line exhibits two mutually exclusive responses to FGF-1 and EGF, entering mitosis at cell confluency while undergoing an epithelium-to-mesenchyme transition (EMT) when cultured at subconfluency. EMT is characterized by acquisition of cell motility, modifications of cell morphology, and cell dissociation correlating with the loss of desmosomes from cellular cortex. The pleiotropic effects of EGF and FGF-1 on NBT-II cells suggest that multiple signaling pathways may be activated. We demonstrate here that growth factor activation is linked to at least two intracellular signaling pathways. One pathway leading to EMT involves an early and sustained stimulation of pp60c-src kinase activity, which is not observed during the growth factor-induced entry into the cell cycle. Overexpression of normal c-src causes a subpopulation of cells to undergo spontaneous EMT and sensitizes the rest of the population to the scattering activity of EGF and FGF-1 without affecting their mitogenic responsiveness. Addition of cholera toxin, a cAMP-elevating agent, severely perturbs growth factor induction of EMT without altering pp60c-src activation, therefore demonstrating that cAMP blockade takes place downstream or independently of pp60c-src. On the other hand, overexpression of a mutated, constitutively activated form of pp60c-src does not block cell dispersion while strongly inhibiting growth factor-induced entry into cell division. Moreover, stable transfection of a dominant negative mutant of c-src inhibits the scattering response without affecting mitogenesis induced by the growth factors. Altogether, these results suggest a role for pp60c-src in epithelial cell scattering and indicate that pp60c-src might contribute unequally to the two separate biological activities engendered by a single signal.


Assuntos
Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Animais , Movimento Celular/fisiologia , Fator de Crescimento Epidérmico/farmacologia , Fatores de Crescimento de Fibroblastos/farmacologia , Expressão Gênica/fisiologia , Substâncias de Crescimento/fisiologia , Mutação/fisiologia , Ratos , Transfecção , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia , Neoplasias da Bexiga Urinária
8.
J Cell Biol ; 106(2): 451-9, 1988 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3276718

RESUMO

Transforming growth factor-beta (TGF beta) is a regulator of cellular proliferation which can alter the proteolytic activity of cultured cells by enhancing the secretion of endothelial type plasminogen activator inhibitor and affecting the secretion of plasminogen activators (PAs) in cultured fibroblastic cells. We used the TGF beta-responsive malignant human lung adenocarcinoma cell line A549 to study the relationships between the known TGF beta-induced growth inhibition and the effects of TGF beta on the secretion of PA activity by A549 cells. PA activity was quantitated by caseinolysis assays, and characterized by urokinase mRNA analysis, immunoprecipitation, and zymography assays. PA-inhibitor production was observed in autoradiograms of SDS-polyacrylamide gels and reverse zymography assays. It was found that TGF beta enhanced the production of PA activity by these cells, in accordance with an enhancement of urokinase mRNA levels. A concomitant stimulation of type 1 PA-inhibitor production was also observed in A549 cells in response to TGF beta. In contrast to the observations of A549 cells, TGF beta caused a decrease in the expression of both urokinase and the tissue-type PA mRNA in human embryonic WI-38 lung fibroblasts indicating opposite regulation of the expression of PAs in these cells. The results suggest that TGF beta may play a role in the regulation of the invasive, proteolytically active phenotype of certain lung carcinoma cells.


Assuntos
Peptídeos/farmacologia , Células Tumorais Cultivadas/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Linhagem Celular , Precipitação Química , Cicloeximida/farmacologia , Matriz Extracelular/fisiologia , Glicoproteínas/metabolismo , Humanos , Técnicas Imunológicas , Pulmão/citologia , Inativadores de Plasminogênio , Fatores de Crescimento Transformadores , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
9.
J Cell Biol ; 117(3): 583-93, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1374067

RESUMO

A 40-kD protein kinase C (PKC)epsilon related activity was found to associate with human epithelial specific cytokeratin (CK) polypeptides 8 and 18. The kinase activity coimmunoprecipitated with CK8 and 18 and phosphorylated immunoprecipitates of the CK. Immunoblot analysis of CK8/18 immunoprecipitates using an anti-PKC epsilon specific antibody showed that the 40-kD species, and not native PKC epsilon (90 kD) associated with the cytokeratins. Reconstitution experiments demonstrated that purified CK8 or CK18 associated with a 40-kD tryptic fragment of purified PKC epsilon, or with a similar species obtained from cells that express the fragment constitutively but do not express CK8/18. A peptide pseudosubstrate specific for PKC epsilon inhibited phosphorylation of CK8/18 in intact cells or in a kinase assay with CK8/18 immunoprecipitates. Tryptic peptide map analysis of the cytokeratins that were phosphorylated by purified rat brain PKC epsilon or as immunoprecipitates by the associated kinase showed similar phosphopeptides. Furthermore, PKC epsilon immunoreactive species and CK8/18 colocalized using immunofluorescent double staining. We propose that a kinase related to the catalytic fragment of PKC epsilon physically associates with and phosphorylates cytokeratins 8 and 18.


Assuntos
Isoenzimas/metabolismo , Queratinas/metabolismo , Proteína Quinase C/metabolismo , Citoesqueleto/enzimologia , Imunofluorescência , Humanos , Isoenzimas/imunologia , Queratinas/imunologia , Queratinas/isolamento & purificação , Substâncias Macromoleculares , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fosfopeptídeos/metabolismo , Fosforilação , Proteína Quinase C/imunologia , Proteína Quinase C-épsilon , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Células Tumorais Cultivadas/enzimologia
10.
J Cell Biol ; 140(6): 1535-41, 1998 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-9508784

RESUMO

Stromelysin-3 (ST3; Basset, P., J.P. Bellocq, C. Wolf, I. Stoll, P. Hutin, J.M. Limacher, O.L. Podhajcer, M.P. Chenard, M.C. Rio, P. Chambon. 1990. Nature. 348:699-704) is a matrix metalloproteinase (MMP) expressed in mesenchymal cells located close to epithelial cells, during physiological and pathological tissue remodeling processes. In human carcinomas, high ST3 levels are associated with a poor clinical outcome, suggesting that ST3 plays a role during malignant processes. In this study we report the ST3 gene inactivation by homologous recombination. Although ST3 null mice (ST3-/-) were fertile and did not exhibit obvious alterations in appearance and behavior, the lack of ST3 altered malignant processes. Thus, the suppression of ST3 results in a decreased 7, 12-dimethylbenzanthracene-induced tumorigenesis in ST3-/- mice. Moreover, ST3-/- fibroblasts have lost the capacity to promote implantation of MCF7 human malignant epithelial cells in nude mice (P < 0.008). Finally, we show that this ST3 paracrine function requires extracellular matrix (ECM)-associated growth factors. Altogether, these findings give evidence that ST3 promotes, in a paracrine manner, homing of malignant epithelial cells, a key process for both primary tumors and metastases. Therefore, ST3 represents an appropriate target for specific MMP inhibitor(s) in future therapeutical approaches directed against the stromal compartment of human carcinomas.


Assuntos
Células Epiteliais/enzimologia , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Comunicação Parácrina/fisiologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Neoplasias da Mama , Testes de Carcinogenicidade , Carcinógenos , Clonagem Molecular , Matriz Extracelular/fisiologia , Feminino , Fibroblastos/citologia , Fibroblastos/enzimologia , Humanos , Botões de Extremidades/citologia , Masculino , Metaloproteinase 11 da Matriz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Nus , Transplante de Neoplasias , Fenótipo , Gravidez , Recombinação Genética , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia
11.
J Cell Biol ; 131(6 Pt 1): 1387-401, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8522599

RESUMO

The transfer of newly synthesized membrane proteins moving from the rough endoplasmic reticulum (RER) to the Golgi complex has been studied by electron microscopy in HEp-2 cells transfected with cDNAs for chimeric proteins. These proteins consist of a reporter enzyme, horseradish peroxidase (HRP), anchored to the transmembrane domains of two integral membrane proteins, the transferrin receptor and sialyl-transferase. The chimeras are distributed throughout the nuclear envelope, RER, vesicular tubular clusters (VTCs) and a network of tubules in the cis-Golgi area. At 20 degrees C tubules containing chimera connect the RER to the VTCs and to the cis-Golgi network. On transfer to 37 degrees C in the presence of dithiothreitol (DTT), the chimeras are seen to move from the RER and through the Golgi stack. With this temperature shift the direct connections with the RER are lost and free vesicles form; some of these vesicles contain HRP reaction product which is much more concentrated than in the adjacent RER while others lack reaction product entirely. In cells expressing SSHRPKDEL, DAB reaction product remains distributed throughout the RER, the VTCs, and the cis-Golgi network for prolonged periods in the presence of DTT and almost all of the vesicles which form at 37 degrees C are DAB-positive. Together these observations demonstrate that all three chimeras are transported from the RER to the cis-Golgi in free, 40-60-nm vesicles at 37 degrees C. They also suggest that the retrograde traffic which carries SSHRPKDEL back to the RER is probably mediated by vesicles with a similar morphology but which, in cells expressing membrane-anchored chimeras, lack detectable reaction product.


Assuntos
Retículo Endoplasmático Rugoso/metabolismo , Complexo de Golgi/metabolismo , Sinais Direcionadores de Proteínas , Sequência de Aminoácidos , Sequência de Bases , Transporte Biológico/fisiologia , Compartimento Celular/fisiologia , Ditiotreitol/farmacologia , Retículo Endoplasmático Rugoso/ultraestrutura , Exocitose/fisiologia , Complexo de Golgi/ultraestrutura , Peroxidase do Rábano Silvestre , Humanos , Neoplasias Laríngeas , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Microscopia Eletrônica , Dados de Sequência Molecular , Oligopeptídeos/metabolismo , Receptores da Transferrina/biossíntese , Receptores da Transferrina/metabolismo , Receptores da Transferrina/ultraestrutura , Proteínas Recombinantes de Fusão/metabolismo , Sialiltransferases/metabolismo , Temperatura , Células Tumorais Cultivadas/enzimologia , p-Dimetilaminoazobenzeno
12.
J Cell Biol ; 114(3): 577-83, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1860886

RESUMO

We present a new human osteosarcoma cell line designated OHS-4. These cells showed a high alkaline phosphatase activity that is not regulated by 1,25 dihydroxyvitamin D3. They exhibited a sensitive adenylate cyclase response to parathyroid hormone but not to prostaglandin E2 or human calcitonin. By Northern blot analysis we could detect type I collagen mRNA but none for type III collagen. The cells were able to produce human osteocalcin at a maximum level of 35 ng per million cells when exposed to 2.4 nM 1,25-dihydroxyvitamin D3 for 96 h. We purified this protein from conditioned media using successive chromatography and assessed its identity by partial amino acid sequencing. When injected into nude mice, the cells retained their osteogenic activity and developed calcified tumors. After Von Kossa staining, we observed nonmineralized osteoid deposits and mineralized deposits with a structure similar to that of trabecular bone by light microscopy. On the basis of its osteoblastic characteristics, this new osteosarcoma cell line may represent the human counterpart of the ROS 17/2 cell line. This cell line represents a valuable model for the isolation and characterization of human bone specific proteins.


Assuntos
Osteossarcoma , Células Tumorais Cultivadas , Adenilil Ciclases/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Calcitriol/farmacologia , Divisão Celular , Cromatografia Líquida de Alta Pressão , Células Clonais , Colágeno/biossíntese , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Osteocalcina/isolamento & purificação , Osteocalcina/metabolismo , Osteossarcoma/enzimologia , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Hormônio Paratireóideo/farmacologia , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/metabolismo
13.
J Cell Biol ; 138(6): 1379-94, 1997 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-9298992

RESUMO

Keratins 8 (K8) and 18 (K18) are major components of intermediate filaments (IFs) of simple epithelial cells and tumors derived from such cells. Structural cell changes during apoptosis are mediated by proteases of the caspase family. During apoptosis, K18 IFs reorganize into granular structures enriched for K18 phosphorylated on serine 53. K18, but not K8, generates a proteolytic fragment during drug- and UV light-induced apoptosis; this fragment comigrates with K18 cleaved in vitro by caspase-6, -3, and -7. K18 is cleaved by caspase-6 into NH2-terminal, 26-kD and COOH-terminal, 22-kD fragments; caspase-3 and -7 additionally cleave the 22-kD fragment into a 19-kD fragment. The cleavage site common for the three caspases was the sequence VEVD/A, located in the conserved L1-2 linker region of K18. The additional site for caspases-3 and -7 that is not cleaved efficiently by caspase-6 is located in the COOH-terminal tail domain of K18. Expression of K18 with alanine instead of serine at position 53 demonstrated that cleavage during apoptosis does not require phosphorylation of serine 53. However, K18 with a glutamate instead of aspartate at position 238 was resistant to proteolysis during apoptosis. Furthermore, this cleavage site mutant appears to cause keratin filament reorganization in stably transfected clones. The identification of the L1-2 caspase cleavage site, and the conservation of the same or very similar sites in multiple other intermediate filament proteins, suggests that the processing of IFs during apoptosis may be initiated by a similar caspase cleavage.


Assuntos
Apoptose/fisiologia , Caspases , Cisteína Endopeptidases/metabolismo , Filamentos Intermediários/metabolismo , Queratinas/metabolismo , Adenocarcinoma , Animais , Caspase 3 , Caspase 6 , Caspase 7 , Neoplasias do Endométrio , Precursores Enzimáticos/metabolismo , Epitélio/química , Epitélio/enzimologia , Feminino , Expressão Gênica/fisiologia , Humanos , Queratinas/química , Queratinas/genética , Camundongos , Dados de Sequência Molecular , Mutagênese/fisiologia , Mapeamento de Peptídeos , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia
14.
J Cell Biol ; 151(5): 951-9, 2000 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-11085998

RESUMO

During apoptosis, caspases, a family of proteases, disassemble a cell by cleaving a set of proteins. Caspase-3 plays a major role in the dissassembly of the nucleus by processing several nuclear substrates. The question is how caspase-3 which is usually cytoplasmic, gains access to its nuclear targets. It was suggested that caspase-3 is actively transported to the nucleus through the nuclear pores. We found that caspase-9, which is activated earlier than caspase-3, directly or indirectly inactivates nuclear transport and increases the diffusion limit of the nuclear pores. This increase allows caspase-3 and other molecules that could not pass through the nuclear pores in living cells to enter or leave the nucleus during apoptosis by diffusion. Hence, caspase-9 contributes to cell disassembly by disrupting the nuclear cytoplasmic barrier.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Citoplasma/enzimologia , Proteínas do Tecido Nervoso/genética , Poro Nuclear/enzimologia , Espectrina/genética , Transporte Ativo do Núcleo Celular/fisiologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama , Caspase 3 , Caspase 8 , Caspase 9 , Compartimento Celular/fisiologia , Cisplatino/farmacologia , Difusão , Feminino , Humanos , Proteínas do Tecido Nervoso/química , Espectrina/química , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia
15.
J Cell Biol ; 131(1): 227-34, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7559779

RESUMO

Studies on the molecular mechanisms underlying neuronal differentiation are frequently performed using cell lines established from neuroblastomas. In this study we have used mouse N1E-115 neuroblastoma cells that undergo neuronal differentiation in response to DMSO. During differentiation, cyclin-dependent kinase (cdk) activities decline and phosphorylation of the retinoblastoma gene product (pRb) is lost, leading to the appearance of a pRb-containing E2F DNA-binding complex. The loss of cdk2 activity is due to a decrease in cdk2 abundance whereas loss of cdk4 activity is caused by strong association with the cdk inhibitor (CKI) p27KIP1 and concurrent loss of cdk4 phosphorylation. Moreover, neuronal differentiation can be induced by overexpression of p27KIP1 or pRb, suggesting that inhibition of cdk activity leading to loss of pRb phosphorylation, is the major determinant for neuronal differentiation.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Transporte , Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas de Ligação a DNA , Neuroblastoma/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas , Proteínas Supressoras de Tumor , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Fatores de Transcrição E2F , Inibidores Enzimáticos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Neuroblastoma/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas/enzimologia
16.
J Cell Biol ; 131(1): 235-42, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7559780

RESUMO

Proliferation of some cultured human tumor cell lines bearing high numbers of epidermal growth factor (EGF) receptors is paradoxically inhibited by EGF in nanomolar concentrations. In the present study, we have investigated the biochemical mechanism of growth inhibition in A431 human squamous carcinoma cells exposed to exogenous EGF. In parallel, we studied a selected subpopulation, A431-F, which is resistant to EGF-mediated growth inhibition. We observed a marked reduction in cyclin-dependent kinase-2 (CDK2) activity when A431 and A431-F cells were cultured with 20 nM EGF for 4 h. After further continuous exposure of A431 cells to EGF, the CDK2 activity remained at a low level and was accompanied by persistent G1 arrest. In contrast, the early reduced CDK2 activity and G1 accumulation in A431-F cells was only transient. We found that, at early time points (4-8 h), EGF induces p21Cip1/WAF1 mRNA and protein expression in both EGF-sensitive A431 cells and EGF-resistant A431-F cells. But only in A431 cells, was p21Cip1/WAF1 expression sustained at a significantly increased level for up to 5 d after addition of EGF. Induction of p21Cip1/WAF1 by EGF could be inhibited by a specific EGF receptor tyrosine kinase inhibitor, tyrphostin AG1478, suggesting that p21Cip1/WAF1 induction was a consequence of receptor tyrosine kinase activation by EGF. We also demonstrated that the increased p21Cip1/WAF1 was associated with both CDK2 and proliferating cell nuclear antigen (PCNA). Taken together, our results demonstrate that p21Cip1/WAF1 is an important mediator of EGF-induced G1 arrest and growth inhibition in A431 cells.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Inibidores Enzimáticos/metabolismo , Receptores ErbB/fisiologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Tirfostinas , Carcinoma de Células Escamosas , Ciclo Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Fator de Crescimento Epidérmico/farmacologia , Inibidores do Crescimento/fisiologia , Humanos , Nitrilas/farmacologia , Fenóis/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas/enzimologia
17.
J Cell Biol ; 137(5): 1103-16, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9166410

RESUMO

The alpha-catenin molecule links E-cadherin/ beta-catenin or E-cadherin/plakoglobin complexes to the actin cytoskeleton. We studied several invasive human colon carcinoma cell lines lacking alpha-catenin. They showed a solitary and rounded morphotype that correlated with increased invasiveness. These round cell variants acquired a more normal epithelial phenotype upon transfection with an alpha-catenin expression plasmid, but also upon treatment with the protein kinase C (PKC) activator 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Video registrations showed that the cells started to establish elaborated intercellular junctions within 30 min after addition of TPA. Interestingly, this normalizing TPA effect was not associated with alpha-catenin induction. Classical and confocal immunofluorescence showed only minor TPA-induced changes in E-cadherin staining. In contrast, desmosomal and tight junctional proteins were dramatically rearranged, with a conversion from cytoplasmic clusters to obvious concentration at cell-cell contacts and exposition at the exterior cell surface. Electron microscopical observations revealed the TPA-induced appearance of typical desmosomal plaques. TPA-restored cell-cell adhesion was E-cadherin dependent as demonstrated by a blocking antibody in a cell aggregation assay. Addition of an antibody against the extracellular part of desmoglein-2 blocked the TPA effect, too. Remarkably, the combination of anti-E-cadherin and anti-desmoglein antibodies synergistically inhibited the TPA effect. Our studies show that it is possible to bypass the need for normal alpha-catenin expression to establish tight intercellular adhesion by epithelial cells. Apparently, the underlying mechanism comprises upregulation of desmosomes and tight junctions by activation of the PKC signaling pathway, whereas E-cadherin remains essential for basic cell-cell adhesion, even in the absence of alpha-catenin.


Assuntos
Proteínas do Citoesqueleto/deficiência , Desmossomos/química , Desmossomos/enzimologia , Proteína Quinase C/metabolismo , Transativadores , Antígenos de Superfície/metabolismo , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Tamanho Celular/fisiologia , Neoplasias do Colo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , DNA Complementar , Detergentes , Humanos , Microscopia Eletrônica , Solubilidade , Acetato de Tetradecanoilforbol/farmacologia , Junções Íntimas/química , Junções Íntimas/enzimologia , Transfecção , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/ultraestrutura , alfa Catenina , beta Catenina
18.
J Cell Biol ; 138(6): 1219-28, 1997 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-9298978

RESUMO

The oncogene bcl-2 encodes a 26-kD protein localized to intracellular membranes, including the ER, mitochondria, and perinuclear membrane, but its mechanism of action is unknown. We have been investigating the hypothesis that Bcl-2 regulates the movement of calcium ions (Ca2+) through the ER membrane. Earlier findings in this laboratory indicated that Bcl-2 reduces Ca2+ efflux from the ER lumen in WEHI7.2 lymphoma cells treated with the Ca2+-ATPase inhibitor thapsigargin (TG) but does not prevent capacitative entry of extracellular calcium. In this report, we show that sustained elevation of cytosolic Ca2+ due to capacitative entry is not required for induction of apoptosis by TG, suggesting that ER calcium pool depletion may trigger apoptosis. Bcl-2 overexpression maintains Ca2+ uptake in the ER of TG-treated cells and prevents a TG-imposed delay in intralumenal processing of the endogenous glycoprotein cathepsin D. Also, Bcl-2 overexpression preserves the ER Ca2+ pool in untreated cells when extracellular Ca2+ is low. However, low extracellular Ca2+ reduces the antiapoptotic action of Bcl-2, suggesting that cytosolic Ca2+ elevation due to capacitative entry may be required for optimal ER pool filling and apoptosis inhibition by Bcl-2. In summary, the findings suggest that Bcl-2 maintains Ca2+ homeostasis within the ER, thereby inhibiting apoptosis induction by TG.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/química , Homeostase/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cálcio/farmacocinética , ATPases Transportadoras de Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Retículo Endoplasmático/enzimologia , Inibidores Enzimáticos/farmacologia , Linfoma , Camundongos , Tapsigargina/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia
19.
J Cell Biol ; 137(5): 1057-68, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9166406

RESUMO

As a rule, hepatocyte growth factor/scatter factor (HGF/SF) is produced by mesenchymal cells, while its receptor, the tyrosine kinase encoded by the met proto-oncogene, is expressed by the neighboring epithelial cells in a canonical paracrine fashion. In the present work we show that both HGF/SF and met are coexpressed by undifferentiated C2 mouse myoblasts. In growing cells, the autocrine loop is active as the receptor exhibits a constitutive phosphorylation on tyrosine that can be abrogated by exogenously added anti-HGF/SF neutralizing antibodies. The transcription of HGF/SF and met genes is downregulated when myoblasts stop proliferating and differentiate. The coexpression of HGF/SF and met genes is not exclusive to C2 cells since it has been assessed also in other myogenic cell lines and in mouse primary satellite cells, suggesting that HGF/SF could play a role in muscle development through an autocrine way. To analyze the biological effects of HGF/SF receptor activation, we stably expressed the constitutively activated receptor catalytic domain (p65(tpr-met)) in C2 cells. This active kinase determined profound changes in cell shape and inhibited myogenesis at both morphological and biochemical levels. Notably, a complete absence of muscle regulatory markers such as MyoD and myogenin was observed in p65(tpr-met) highly expressing C2 clones. We also studied the effects of the ectopic expression of human isoforms of met receptor (h-met) and of HGF/SF (h-HGF/SF) in stable transfected C2 cells. Single constitutive expression of h-met or h-HGF/SF does not alter substantially the growth and differentiation properties of the myoblast cells, probably because of a species-specific ligand-receptor interaction. A C2 clone expressing simultaneously both h-met and h-HGF/SF is able to grow in soft agar and shows a decrease in myogenic potential comparable to that promoted by p65(tpr-met) kinase. These data indicate that a met kinase signal released from differentiation-dependent control provides a negative stimulus for the onset of myogenic differentiation.


Assuntos
Fator de Crescimento de Hepatócito/farmacologia , Músculos/citologia , Fosfotransferases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Diferenciação Celular/fisiologia , Cães , Regulação para Baixo/fisiologia , Ativação Enzimática , Expressão Gênica/fisiologia , Gliceraldeído-3-Fosfato Desidrogenases/genética , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Túbulos Renais Distais/citologia , Fígado/citologia , Camundongos , Camundongos Endogâmicos C3H , Músculos/química , Músculos/enzimologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Miogenina/genética , Miogenina/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Teratocarcinoma , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
20.
J Cell Biol ; 143(2): 533-45, 1998 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-9786961

RESUMO

We have established a cell culture system that reproduces morphogenic processes in the developing mammary gland. EpH4 mouse mammary epithelial cells cultured in matrigel form branched tubules in the presence of hepatocyte growth factor/scatter factor (HGF/SF), the ligand of the c-met tyrosine kinase receptor. In contrast, alveolar structures are formed in the presence of neuregulin, a ligand of c-erbB tyrosine kinase receptors. These distinct morphogenic responses can also be observed with selected human mammary carcinoma tissue in explant culture. HGF/SF-induced branching was abrogated by the PI3 kinase inhibitors wortmannin and LY294002. In contrast, neuregulin- induced alveolar morphogenesis was inhibited by the MAPK kinase inhibitor PD98059. The c-met-mediated response could also be evoked by transfection of a c-met specific substrate, Gab1, which can activate the PI3 kinase pathway. An activated hybrid receptor that contained the intracellular domain of c-erbB2 receptor suffices to induce alveolar morphogenesis, and was observed in the presence of tyrosine residues Y1028, Y1144, Y1201, and Y1226/27 in the substrate-binding domain of c-erbB2. Our data demonstrate that c-met and c-erbB2 signaling elicit distinct morphogenic programs in mammary epithelial cells: formation of branched tubules relies on a pathway involving PI3 kinase, whereas alveolar morphogenesis requires MAPK kinase.


Assuntos
Glândulas Mamárias Animais/embriologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptor ErbB-2/fisiologia , Transdução de Sinais/fisiologia , Androstadienos/farmacologia , Animais , Neoplasias da Mama , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Moléculas de Adesão Celular/fisiologia , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Glicoproteínas/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/enzimologia , Camundongos , Microscopia Eletrônica , Morfogênese/fisiologia , Morfolinas/farmacologia , Neurregulinas , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/ultraestrutura , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA