Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pain ; 90(1-2): 113-25, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11166977

RESUMO

In the adult brain, neurotrophins play a key role in adaptive processes linked to increased neuronal activity. A growing body of evidence suggests that chronic pain results from long-term plasticity of central pathways involved in nociception. We have investigated the involvement of nerve growth factor (NGF) in adaptive responses of primary sensory neurons during the course of a long-lasting inflammatory pain model. The amount and distribution of the NGF receptors p75(NTR) and TrkA were measured in the dorsal horn and dorsal root ganglia (DRG) of animals subjected to Freund's adjuvant-induced arthritis (AIA). We observed an increased immunoreactivity of both receptors in the central terminals of primary sensory neurons in the arthritic state. The increases were seen in the same population of afferent terminals in deep dorsal horn laminae. These changes paralleled the variations of clinical and behavioral parameters that characterize the course of the disease. They occurred in NGF-sensitive, but not GDNF-sensitive, nerve terminals. However, p75(NTR) and TrkA protein levels in the DRG (in the cell body of these neurons) showed different response patterns. An immediate rise of p75(NTR) was seen in parallel with the initial inflammation that developed after administration of Freund's adjuvant in hindpaws. In contrast, increases of the mature (gp140(trk)) form of TrkA occurred later and seemed to be linked to the development of the long-lasting inflammatory response. The changes in receptor expression were observed exclusively at lumbar levels, L3-L5, somatotopically appropriate for the inflammation. Together, these results implicate NGF in long-term mechanisms accompanying chronic inflammatory pain, via the up-regulation of its high affinity receptor, and offer additional evidence for differential processes underlying short- versus long-lasting inflammatory pain.


Assuntos
Artrite Experimental/metabolismo , Gânglios Espinais/metabolismo , Células do Corno Posterior/metabolismo , Receptor trkA/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Masculino , Neurônios Aferentes/metabolismo , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Fator de Crescimento Neural , Substância P/metabolismo
2.
Eur J Cell Biol ; 79(11): 816-23, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11139145

RESUMO

Spermatogenesis is a highly ordered process which requires mitotic and meiotic divisions. In this work, we studied the relative changes in the levels of the two components of the M-phase promoting factor (MPF): the regulatory subunit cyclin B1 (CycB1) and its catalytic subunit cdk1, in spermatogenic cells of rats between 16 and 90 days of life. A multivariate flow cytometry analysis of forward scatter (FSC), side scatter (SSC) and DNA content was used to identify six populations of rat germ cells: spermatogonia with preleptotene spermatocytes, young pachytene spermatocytes, middle to late pachytene spermatocytes, secondary spermatocytes with doublets of round spermatids, round spermatids, and elongated spermatids. For any population studied no significant difference in the relative cellular content of CycB1 or cdk1 proteins between animals of different ages was observed. By contrast, CycB1 and cdk1 levels were different between the different populations of germ cells. CycB1 and cdk1 were rather high in young pachytene spermatocytes and culminated in late spermatocytes, i.e. just before the first meiotic division. The relative levels of the two proteins remained high in secondary spermatocytes then decreased in round spermatids at the exit of meiosis. Similar results were obtained by Western-blot analysis of total proteins obtained from lysates of elutriated fractions of spermatocytes and spermatids. MPF activity was assessed in lysates of germ cells from 32-day-old rats or adult animals using p13suc1 agarose and histone H1 as an exogenous substrate. H1 kinase activity was higher in pachytene spermatocytes than in round spermatid fractions from both adult and young rats. These results indicate that the meiotic G2/M transition is associated to high levels of CycB1 and cdk1 leading to high MPF activity irrespective of the age of the animals.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Espermátides/metabolismo , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Testículo/citologia , Animais , Ciclo Celular/fisiologia , Ciclina B1 , Citometria de Fluxo , Immunoblotting , Masculino , Ratos , Ratos Sprague-Dawley , Espermátides/crescimento & desenvolvimento , Espermatócitos/crescimento & desenvolvimento , Testículo/metabolismo
3.
J Virol ; 73(10): 8762-70, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10482630

RESUMO

Human erythroid progenitor cells are the main target cells of the human parvovirus B19 (B19), and B19 infection induces a transient erythroid aplastic crisis. Several authors have reported that the nonstructural protein 1 (NS-1) encoded by this virus has a cytotoxic effect, but the underlying mechanism of NS-1-induced primary erythroid cell death is still not clear. In human erythroid progenitor cells, we investigated the molecular mechanisms leading to apoptosis after natural infection of these cells by the B19 virus. The cytotoxicity of NS-1 was concomitantly evaluated in transfected erythroid cells. B19 infection and NS-1 expression induced DNA fragmentation characteristic of apoptosis, and the commitment of erythroid cells to undergo apoptosis was combined with their accumulation in the G(2) phase of the cell cycle. Since B19- and NS-1-induced apoptosis was inhibited by caspase 3, 6, and 8 inhibitors, and substantial caspase 3, 6, and 8 activities were induced by NS-1 expression, there may have been interactions between NS-1 and the apoptotic pathways of the death receptors tumor necrosis factor receptor 1 and Fas. Our results suggest that Fas-FasL interaction was not involved in NS-1- or B19-induced apoptosis in erythroid cells. In contrast, these cells were sensitized to tumor necrosis factor alpha (TNF-alpha)-induced apoptosis. Moreover, the ceramide level was enhanced by B19 infection and NS-1 expression. Therefore, our results suggest that there may be a connection between the respective apoptotic pathways activated by TNF-alpha and NS-1 in human erythroid cells.


Assuntos
Apoptose , Eritrócitos/patologia , Eritrócitos/virologia , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Parvovirus B19 Humano , Fator de Necrose Tumoral alfa/metabolismo , Proteínas não Estruturais Virais/metabolismo , Eritrócitos/metabolismo , Humanos , Infecções por Parvoviridae/metabolismo , Transdução de Sinais
4.
Biochem Biophys Res Commun ; 262(1): 157-62, 1999 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-10448085

RESUMO

The transcriptional coactivator CBP displays an intrinsic histone acetyl transferase (HAT) activity which seems to participate in transcriptional activation through the destabilization of nucleosome structure. CBP is involved in the activity of several transcription factors that are nuclear endpoints of intracellular signal transduction pathways. In some instances, the transcription factors are phosphorylated upon cell activation, which induces their interaction with CBP. CBP itself is a phosphoprotein and can be phosphorylated by cycle-dependent kinases or by MAP kinases. Here we show that CBP phosphorylation by p44 MAP kinase/ERK1 results in the stimulation of its HAT enzymatic activity. The p44 MAP kinase/ERK1 phosphorylation sites are located in the C-terminal part of the protein, outside of the HAT domain. These sites are required for enzymatic stimulation, suggesting that phosphorylation by p44 MAP kinase/ERK1 induces a conformational change of the CBP molecule. Our data suggest that, in some instances, CBP itself might be a target for signal transduction pathways.


Assuntos
Acetiltransferases/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae , Transativadores/metabolismo , Células 3T3 , Acetiltransferases/química , Trifosfato de Adenosina/metabolismo , Animais , Proteína de Ligação a CREB , Ativação Enzimática , Histona Acetiltransferases , Histonas/metabolismo , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Modelos Genéticos , Proteínas Nucleares/química , Nucleossomos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Transdução de Sinais , Fatores de Tempo , Transativadores/química
5.
Oncogene ; 18(18): 2872-82, 1999 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-10362258

RESUMO

Addition of nerve growth factor (NGF) to PC12 cells promotes neuronal differentiation while inhibiting cell proliferation. In order to understand how NGF exerts its antimitogenic effect during differentiation, we have studied the mechanism by which this factor activates the promoter of the CDK inhibitor p21W4F1/CIP1. The minimal region of the p21 promoter required for the NGF-induction was mapped to a contiguous stretch of 10 bp located 83 bases upstream of the transcription initiation site. This GC-rich region was shown to interact specifically with the transcription factor Sp1 and the related protein Sp3, in either exponentially-growing or NGF-treated PC12 cells. The addition of NGF resulted in an accumulation of the transcriptional co-activator p300 in complexes associated with the NGF-responsive region. Transcriptional activity of Sp1, Sp3 and p300 was specifically induced by NGF in a Gal4-fusion assay, indicating that induction of p21 during neuronal differentiation may involve regulation of the activity of these factors by NGF. Furthermore, p300 was able to act as a co-activator for Sp1-mediated transcriptional activation in PC12 cells, suggesting that p300 and Sp1 may cooperate in activating p21 transcription during the withdrawal of neuronal precursors from the cell cycle. This hypothesis is supported by experiments showing that p300 and Sp1 form complexes in PC12 cells.


Assuntos
Ciclinas/genética , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Fator de Transcrição Sp1/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína p300 Associada a E1A , Galactosidases/genética , Galactosidases/metabolismo , Humanos , Fatores de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células PC12/metabolismo , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição Sp3 , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
6.
Hum Gene Ther ; 10(7): 1129-38, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10340545

RESUMO

Retroviral vectors allow stable integration of exogenous DNA into genomic DNA and therefore gene transmission to progeny. Multipotent neural precursors and immortal cell lines prepared from them have been demonstrated to integrate into either adult or developing brain in a nontumorigenic, functional manner, without interfering with normal neurobiological processes. These cells thus appear to provide a Trojan horse ideally adapted to directing the expression of transgenes appropriately in a host brain. Here we investigated and optimized the transduction capacity of MuLV-based retroviral vectors in which internal ribosomal entry segments (IRESs) drive coexpression of two heterologous gene products from a single bicistronic mRNA in a human multipotent neural precursor cell line, "Dev," which was prepared from a medulloblastoma. For this, two vectors containing two different combinations of three viral IRESs were used and the capacity of different pseudotyped vectors to permit an efficient and stable transduction of Dev cells was compared. Our data show that (1) the best recombinant vectors for Dev cell transduction are those pseudotyped with the 10A1 MuLV envelope (40% of transduction) and (2) the initial coexpression of neo and plap, observed in transduced undifferentiated Dev cells, is maintained in differentiated Dev cells with a neuronal or glial phenotype. Therefore, these double-IRES vectors may provide an efficient means of transducing the coexpression of two proteins in undifferentiated human neural precursors that is maintained in their differentiated progeny. These data suggest that the double-IRES strategy is well adapted to potential therapeutic situations when coexpression of two different transgenes may be required in the same cell.


Assuntos
Vetores Genéticos , Neuroglia/metabolismo , Neurônios/metabolismo , Retroviridae/genética , Animais , Diferenciação Celular , Humanos , Canamicina Quinase/metabolismo , Meduloblastoma , Camundongos , Neuroglia/citologia , Neurônios/citologia , Ribossomos/metabolismo , Transdução Genética , Células Tumorais Cultivadas
7.
Neuroreport ; 9(15): 3409-13, 1998 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-9855290

RESUMO

Retinal ganglion cells of the fish have the spontaneous capacity to regenerate after nerve crush, a phenomenon known to be facilitated by nerve growth factor (NGF). We have studied the high-affinity NGF receptor TrkA, during the regeneration of the tench (Tinca tinca L.) optic nerve, using immunocytochemical techniques. TrkA-like immunoreactivity increased during the regeneration of the retinal ganglion cells. The increase is followed by a change in the subcellular distribution from perinuclear in control cells to cytoplasmic and perinuclear in regenerating ones. This increase was observed when antibodies against the extracellular domain of TrkA were used; no changes in TrkA-like immunoreactivity were observed with antibodies against the intracellular domain of TrkA. We thus conclude that modulation of TrkA is involved in the regeneration of fish retinal ganglion cells.


Assuntos
Regeneração Nervosa/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Células Ganglionares da Retina/química , Células Ganglionares da Retina/citologia , Células 3T3 , Animais , Anticorpos , Especificidade de Anticorpos , Western Blotting , Cyprinidae , Epitopos/análise , Imuno-Histoquímica , Camundongos , Compressão Nervosa , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/imunologia , Receptor trkA , Receptores de Fator de Crescimento Neural/análise , Receptores de Fator de Crescimento Neural/imunologia
8.
Oncogene ; 17(13): 1663-72, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9796695

RESUMO

Primitive neuroectodermal tumours (PNET) are thought to derive from the malignant transformation of pluripotent CNS precursors although this hypothesis has yet to be tested in vitro. Here we show that cells of a human PNET cell line 'Dev' express functional fibroblast growth factor (FGF) receptors (FGFR) and respond to FGF2 as multipotent CNS precursors. FGF2 induces tyrosine phosphorylation of FGFR-1 and FGFR-2 and many cellular substrates including MAP kinases and stimulates proliferation. Cells detach from plastic substrates and proliferate to form large clusters of undifferentiated cells. After adhesion to polylysine, cells migrate out from the clusters and differentiate. The majority of differentiated cells express neuronal phenotypes but distinct subpopulations express oligodendrocytic and astrocytic markers. Mature neural differentiation markers are not otherwise detected in Dev cells in defined medium. Identical results were obtained with 12 monoclonal subclones as well as the parent cell line, confirming that Dev cells are multipotent. This extends evidence that multipotent CNS precursors are the cellular substrate from which certain PNET develop and shows that FGF2 is a potent proliferation and differentiation inducer for PNET cells in vitro, suggesting that FGF2 may also modulate the evolution of PNET in vivo. Finally our results suggest that PNET cell lines may provide models to elucidate the biology of human multipotent CNS precursors.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Tumores Neuroectodérmicos Primitivos , Neurônios/citologia , Biomarcadores , Diferenciação Celular , Divisão Celular , Células Clonais , Meios de Cultura Livres de Soro , Células Epiteliais , Humanos , Neurônios/efeitos dos fármacos , Fenótipo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Células-Tronco , Células Tumorais Cultivadas
9.
J Neurosci ; 18(17): 6767-75, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9712648

RESUMO

Expression of the nerve growth factor (NGF) receptors TrkA and p75(NTR) was found to vary at the surface of PC12 cells in a cell cycle phase-specific manner. This was evidenced by using flow cytometric and microscopic analysis of cell populations labeled with antibodies to the extracellular domains of both receptors. Differential expression of these receptors also was evidenced by biotinylation of surface proteins and Western analysis, using antibodies specific for the extracellular domains of TrkA and p75(NTR). TrkA is expressed most strongly at the cell surface in M and early G1 phases, whereas p75(NTR) is expressed mainly in late G1, S, and G2 phases. This expression reflects the molecular and cellular responses to NGF in specific phases of the cell cycle; in the G1 phase NGF elicits both the anti-mitogenic effect, i.e., inhibition of the G1 to S transition, and the differentiation response whereas a survival effect is provoked elsewhere in the cell cycle. A model is proposed relating these responses to the surface expression of the two receptors. These observations open the way for novel approaches to the investigation of the mechanism of NGF signal transduction.


Assuntos
Ciclo Celular/fisiologia , Proteínas Proto-Oncogênicas/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Receptores de Fator de Crescimento Neural/biossíntese , Animais , Biotinilação , Células PC12 , Ratos , Receptor de Fator de Crescimento Neural , Receptor trkA
10.
Oncogene ; 16(7): 825-32, 1998 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-9484773

RESUMO

Survival signalling by ligand-activated tyrosine kinase receptors plays a crucial role in maintaining the balance between cell viability and apoptosis in multicellular organisms. To identify receptor domains and pathways involved in survival signalling, the nerve growth factor receptor TrkA was expressed in Rat-1/MycER fibroblasts. We demonstrate that wt-TrkA receptor delays c-Myc-, U.V.- and Cycloheximide-induced apoptosis and activates targets such as the mitogen-activated protein kinase (MAPK) Erk2 and the serine/threonine kinase Akt/PKB, both of which have been implicated in survival signalling. TrkA mutated within its SHC binding site (Y490F) delays c-Myc-induced apoptosis without activating endogenous Akt/PKB. In contrast, the TrkA Y490F mutant receptor does not delay U.V.-induced apoptosis whilst TrkA mutated at its PLC-gamma binding site (Y785F) is capable of protecting from apoptosis induced by c-Myc or U.V. treatment. The double mutant TrkA YY490/785FF fails to block either of these two apoptotic stimuli. While P13-kinase inhibitors LY294002 and Wortmannin completely block survival signalling following U.V. treatment, neither drug affects the ability of TrkA to block c-Myc-induced apoptosis. We show that the Akt/PKB pathway is essential for NGF stimulated TrkA survival signalling in the case of U.V.-induced apoptosis, but that apoptosis induced by c-Myc is also blocked by a novel, Akt/PKB-independent, pathway. These observations suggest that TrkA can activate different survival signalling pathways, which can interfere with specific apoptotic pathways.


Assuntos
Apoptose , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Sobrevivência Celular , Ativação Enzimática , Genes myc , Humanos , Proteína Quinase 1 Ativada por Mitógeno , Mutagênese Sítio-Dirigida , Mutação Puntual , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Receptor trkA , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Raios Ultravioleta
11.
Oncogene ; 15(4): 393-402, 1997 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-9242375

RESUMO

Multiple endocrine neoplasia type 2A (MEN 2A) and familial medullary thyroid carcinoma (FMTC) are two dominantly inherited disorders caused by germline mutations of the RET proto-oncogene. The RET gene codes for a receptor tyrosine kinase. The majority of MEN2A and FMTC mutations are clustered in the extra-cellular cysteine-rich domain and result in constitutive activation of the tyrosine kinase through the formation of disulfide-bonded RET homodimers. Recently, two novel point mutations have been identified in the germline of five distinct FMTC families. Both mutations occur within the catalytic domain of the RET kinase and lead to the substitution of either glutamic acid 768 or valine 804 by an aspartic acid and a leucine respectively. We have introduced each FMTC mutation in two RET isoforms: RET51 the long isoform (1114 aa) and RET9 the short isoform (1072 aa) which differ in the C-terminal region of the protein. The RET51 isoform carrying either E768D or V804L mutation was autophosphorylated, displayed a transforming activity upon expression in Rat1 fibroblasts and induced neuronal differentiation of PC12 cells. However, the transforming capacity of these RET51-FMTC mutants was found to be severalfold less potent compared to the same isoform carrying either the MEN2A mutation (C634R) or the MEN2B mutation (M918T). In contrast, RET9 containing mutations E768D or V804L was not autophosphorylated, exhibited a poor oncogenic potential in fibroblasts and did not promote neuritic outgrowth upon expression in PC12 cells. Overall, these findings demonstrate that mutations E768D and V804L are gain-of-function mutations that confer to the long RET isoform the capacity to exert a biological effect, although these mutations are more weakly activating than the MEN2A and MEN2B mutations. These results may provide a biochemical basis as to why the phenotypic consequences of these mutations are restricted to thyroid C-cells.


Assuntos
Carcinoma Medular/genética , Transformação Celular Neoplásica , Proteínas de Drosophila , Mutação Puntual , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias da Glândula Tireoide/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Diferenciação Celular , Ativação Enzimática , Camundongos , Dados de Sequência Molecular , Células PC12 , Proteínas Proto-Oncogênicas c-ret , Ratos
12.
Oncogene ; 14(3): 265-75, 1997 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-9018112

RESUMO

Germline mutations of the RET proto-oncogene, which codes for a receptor tyrosine kinase, cause multiple endocrine neoplasia type 2A (MEN 2A) and 2B (MEN 2B) and familial medullary thyroid carcinoma (FMTC). MEN 2 mutations have been shown to result in RET oncogenic activation. The RET gene encodes several isoforms whose biological properties, when altered by MEN 2 mutations, have not been thoroughly addressed yet. In this study, we have introduced a MEN 2A mutation (Cys634-->Arg) and the unique MEN 2B mutation (Met918-->Thr) in two RET isoforms of 1114 and 1072 amino acids which differ in the carboxy-terminus part. Herein, we report that each RET isoform activated by MEN 2A or MEN 2B mutation was transforming in fibroblasts and induced neuronal differentiation of pheochromocytoma PC12 cells. However, among the different RET-MEN 2 mutants, the long RET isoform activated by the MEN 2B mutation stimulated the most prominent neurite outgrowth in PC12 cells, while the short RET isoform counterpart elicited a very weak differentiation effect in PC12 cells. We further demonstrate that the morphological changes of PC12 cells caused by constitutively activated RET oncoproteins involved the engagement of a Ras-dependent pathway. These findings provide evidence that the biological properties of RET-MEN 2 mutants depend on the interplay between the RET isoforms and the nature of the activating MEN 2 mutation.


Assuntos
Proteínas de Drosophila , Mutação em Linhagem Germinativa/genética , Neoplasia Endócrina Múltipla Tipo 2a/genética , Neoplasia Endócrina Múltipla Tipo 2b/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Diferenciação Celular , Transformação Celular Neoplásica , Ativação Enzimática , Vetores Genéticos/genética , Humanos , Neuritos/patologia , Feocromocitoma/patologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-ret , Proto-Oncogenes/fisiologia , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/fisiologia , Retroviridae/genética , Transfecção
13.
Oncogene ; 13(10): 2047-54, 1996 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-8950971

RESUMO

The block of cell proliferation elicited by the addition of nerve growth factor (NGF) to exponentially-growing PC12 cells results, in part, from the inhibition of cyclin D1-associated kinase activity by p21WAF1/CIP1. NGF treatment of PC12 cells provokes the accumulation of p21 mRNA, due to transcriptional activation of the p21 promoter in a p53-independent manner. Transient expression of a mutated form of the adenovirus E1A protein (E1A dCR2), which retains its capacity to bind the transcriptional co-activator p300, completely abolishes the NGF-mediated stimulation of p21 promoter activity. This phenomenon can be reversed by ectopic expression of p300, suggesting that p300 is necessary for the induction of p21 by NGF. In addition, stable expression of E1A dCR2 in PC12 cells results in the inhibition of the NGF response, i.e. it prevents activation of the p21 promoter, cell cycle arrest, and neuronal differentiation. The signalling pathway from the TrkA receptor via the MAP kinase pathway is not altered in these cells. Together, these data indicate that p300 could play a pivotal role in the triggering of the anti-mitogenic effect of NGF and of neuronal differentiation.


Assuntos
Ciclinas/metabolismo , Fatores de Crescimento Neural/farmacologia , Proteínas Nucleares/fisiologia , Transativadores , Fatores de Transcrição/fisiologia , Transcrição Gênica , Animais , Ciclo Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Proteína p300 Associada a E1A , Vetores Genéticos , Humanos , Luciferases/genética , Luciferases/metabolismo , Células PC12 , Feocromocitoma/metabolismo , Feocromocitoma/patologia , RNA Mensageiro/metabolismo , Ratos , Receptores de Fator de Crescimento Neural/metabolismo , Transfecção , Proteína Supressora de Tumor p53/fisiologia
15.
Oncogene ; 12(6): 1347-56, 1996 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-8649837

RESUMO

PC12 cells treated with nerve growth factor (NGF) undergo a G1 block and differentiate. Expression of selected cell cycle regulatory proteins was studied under culture conditions which permit observation of a differentiation response independently from a mitogenic or anti-mitogenic response. The expression of all cell cycle regulatory proteins studied is modulated by NGF addition to exponentially-growing cultures in the presence of serum. While levels of most of these proteins decrease, accumulation of cyclin D1 and the cyclin-dependent kinase inhibitor p21 Cip1/WAF1 is observed. Cyclin D1 associated kinase activity is inhibited, correlating with an increase in p21 protein. PC12 cells, synchronized by serum starvation, undergo morphological and functional differentiation in the presence of NGF. Neither cyclin D1 nor p21 are present in such cultures, nor is their expression upregulated by NGF, indicating that they are not required for this process. Removal of serum from differentiated PC12 cells results in loss of these proteins, but has no effect on differentiation or the nonproliferative state in presence of NGF. Together, the results indicate that cyclin D1 and p21 are not necessary for differentiation per se, nor are they required for maintenance of the differentiated state in the absence of serum.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Fatores de Crescimento Neural/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Fase G1/efeitos dos fármacos , Expressão Gênica , Células PC12 , Ratos
16.
Oncogene ; 12(4): 855-62, 1996 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8632908

RESUMO

The anti-proliferative effect of nerve growth factor (NGF) on the rat pheochromocytoma cell line PC12 has been previously shown to be accompanied by the accumulation of cells in either the G1 phase with a 2c DNA content, or with a 4c DNA content characteristic for G2/M, as evidenced by flow cytometric analysis of DNA distribution using propidium iodide. Herein, these apparently conflicting results are clarified. The present studies indicate that a simple DNA distribution profile obtained by this technique can confound interpretation of the biological effects of NGF on cell-cycle distribution due to the presence of tetraploid cells. Using cyclin D1 and incorporation of bromodeoxyuridine as markers of respectively, G1 and S phase, we show that PC12 cultures can have a considerable amount of tetraploid cells which, when in the G1 phase, have a 4c DNA content and express cyclin D1. During exposure to NGF, this population increases, reflecting the accumulation of cells in the G1 phase of the cell cycle. The data presented, support the possibility that events affecting the expression or action of G1 regulatory proteins may be involved in the molecular mechanism of the anti-mitogenic effect of NGF.


Assuntos
Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ciclinas/biossíntese , Fatores de Crescimento Neural/farmacologia , Proteínas Oncogênicas/biossíntese , Neoplasias das Glândulas Suprarrenais , Animais , Bromodesoxiuridina , Ciclina D1 , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Citometria de Fluxo , Fase G1/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Cinética , Células PC12 , Feocromocitoma , Ratos , Fase S/efeitos dos fármacos
17.
Oncogene ; 12(2): 309-22, 1996 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-8570208

RESUMO

The expression of E and D-type cyclins, Cyclin-Dependent Kinase (CDK) 2 and 4, as well as CDK inhibitors p21Cip1 and p27Kip1 were examined during in vitro differentiation of mouse embryonic stem (ES) cells. ES cells cultured in presence of Differentiation Inhibitory Activity/Leukemia Inhibitory Factor (DIA/LIF) express very low levels of cyclin E/CDK2 complexes, p21Cip1 and p27Kip1 CDK inhibitors, while cyclin D/CDK4-associated kinase activity is undetectable. Withdrawal of DIA/LIF, which induces differentiation, results in the progressive up-regulation of all. Up-regulation of D cyclins occurs through an increase in the steady-state levels of mRNA, concomitantly with the activation of Brachyury and Goosecoid, two early markers of mesoderm differentiation. Similarly, cells from the epiblast of the early postimplantation mouse embryo do not express any cyclin D/CDK4 complexes. These are progressively upregulated at gastrulation and early organogenesis. DIA/LIF-stimulated ES cells are not growth-arrested by overexpression of p16Ink4a, a specific inhibitor of CDK4 and CDK6. We propose that the G1/S transition may be regulated by a minimal mechanism in mouse embryonic stem cells. Induction of differentiation triggers the establishment of a more sophisticated mechanism involving both cyclin D/CDK4- and CDK inhibitor-associated control of G1-phase progression.


Assuntos
Proteínas de Ciclo Celular , Ciclinas/biossíntese , Embrião de Mamíferos/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores do Crescimento/fisiologia , Interleucina-6 , Linfocinas/fisiologia , Proteínas Associadas aos Microtúbulos/biossíntese , Células-Tronco/metabolismo , Proteínas Supressoras de Tumor , Animais , Sequência de Bases , Proteínas de Transporte/fisiologia , Diferenciação Celular , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Fase G1 , Fator Inibidor de Leucemia , Camundongos , Dados de Sequência Molecular , Regulação para Cima
18.
J Biol Chem ; 269(5): 3739-44, 1994 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-8106419

RESUMO

Nerve growth factor (NGF) stimulates the differentiation of PC12 pheochromocytoma cells to those resembling sympathetic neurons. We have investigated whether NGF regulates transforming growth factor (TGF)-beta gene expression and protein secretion in PC12 cells. These cells constitutively express TGF-beta 1 mRNA, whereas TGF-beta 2 and -beta 3 mRNAs are expressed at very low levels. TGF-beta 1 gene expression was stimulated greater than 10-fold when PC12 cells were treated with NGF. Sequences between -119 and -98 in the TGF-beta 1 promoter, homologous to an Egr-1 binding site, were shown to be important for both basal and NGF-induced promoter activity. We also found that a factor(s) present in nuclear extracts from PC12 cells interacted with the sequences between -119 and -98 and that expression of this factor was induced by NGF treatment. Moreover, specific binding to TGF-beta 1 promoter fragments between -119 and -98 was seen using the bacterially expressed transcription factor Egr-1. These results indicate that activation of TGF-beta 1 expression is one of the cellular responses of PC12 cells to NGF and suggest that TGF-beta may play a role in the differentiation of sympathetic neurons.


Assuntos
Expressão Gênica/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/biossíntese , Neoplasias das Glândulas Suprarrenais , Animais , Sequência de Bases , Sítios de Ligação , Northern Blotting , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/biossíntese , Cloranfenicol O-Acetiltransferase/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Cinética , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Células PC12 , Feocromocitoma , Plasmídeos , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Neoplásico/análise , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Transfecção , Dedos de Zinco
19.
Differentiation ; 54(3): 161-75, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8270144

RESUMO

Nerve growth factor (NGF), a target-derived neurotrophic substance, may have broader biological functions in various types of non-neuronal differentiating cells. The effects of NGF are dependent on initial binding of NGF to specific cell-surface receptors (p75NGFR and p140prototrk) on responsive cells. The continuously growing rat incisor offers an excellent model demonstrating defined territories of differentiation of specific cell populations. We used immunohistochemistry to determine sites of NGF, proNGF and p75NGFR accumulation in the rat incisor, whereas NGF mRNA expression was visualized by in situ hybridization in the developing rat molar and incisor. Strictly similar patterns of NGF mRNA, proNGF and NGF expression were observed in differentiating cells responsible for the production of the main structural matrices of the tooth. Thus, proNGF-like and NGF-like immunoreactivity, as well as the NGF mRNA signal were observed in preameloblasts and young ameloblasts of the dental epithelium and in polarizing odontoblasts of the dental mesenchyme. In contrast, the distribution of p75NGFR was correlated with differentiation event only in dental mesenchyme: polarizing odontoblasts expressed p75NGFR whereas the molecule was absent in functional odontoblasts. In dental epithelium, the restricted expression of p75NGFR in ameloblast precursor cells was correlated with proliferative phenomena. The patterns of proNGF, NGF and p75NGFR expression in epithelium and mesenchyme implicate both an autocrine and paracrine mode of action of the NGF molecule in dental tissues. The findings reported here are important for understanding NGF action in specific dental cell populations and suggest that this molecule is involved in the cascade of events that directs tooth development.


Assuntos
Incisivo/química , Dente Molar/química , Fatores de Crescimento Neural/análise , Precursores de Proteínas/análise , Receptores de Fator de Crescimento Neural/análise , Animais , Diferenciação Celular , Divisão Celular , Feminino , Imuno-Histoquímica , Hibridização In Situ , Incisivo/citologia , Mesoderma/química , Mesoderma/citologia , Dente Molar/citologia , Fatores de Crescimento Neural/genética , Gravidez , Precursores de Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Receptores de Fator de Crescimento Neural/genética
20.
J Neurochem ; 58(1): 175-85, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1309232

RESUMO

Previous studies from this laboratory have shown that the phosphorylation of the S6 protein of the ribosomes is catalyzed by at least two different and separable kinase activities in PC12 cells. One of these activities is increased by treatment of the cells with nerve growth factor, the other by treatment of the cells with epidermal growth factor. The present work shows that these two factors stimulate the phosphorylation of S6 with quite different kinetics, and that both the number of phosphates incorporated into S6 and the phosphopeptide pattern of S6 are different in cells treated with nerve growth factor than in cells treated with epidermal growth factor. The characteristics of the nerve growth factor-sensitive S6 kinase and of the epidermal growth factor-sensitive kinase were also clearly different. Substrate specificity and inhibitor studies indicated that neither was identical to cyclic AMP-dependent kinase, kinase C, or the calcium/calmodulin-dependent kinases. However, two major phosphopeptides produced by S6 phosphorylation in nerve growth factor-treated cells were also seen on phosphorylation of S6 by cyclic AMP-dependent kinase in vitro. In addition, when rat liver 40S ribosomal subunits were pretreated with cyclic AMP-dependent kinase in vitro, the action of the nerve growth factor-sensitive S6 kinase was increased about twofold.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fatores de Crescimento Neural/metabolismo , Células PC12/metabolismo , Proteínas Ribossômicas/metabolismo , Aminoácidos/análise , Animais , Mapeamento de Peptídeos , Fosfopeptídeos/química , Fosforilação , Fosfotransferases/isolamento & purificação , Proteínas Quinases/farmacologia , Proteína S6 Ribossômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA