Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 334
Filtrar
1.
BMC Vet Res ; 13(1): 262, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28830546

RESUMO

BACKGROUND: Spirocerca lupi is a nematode that parasitizes vertebrates in particular canids, by forming nodules in the thoracic cavity specifically in the oesophagus. In 25% of Spirocerca infections of the domestic dog, nodules progress from inflammatory to pre-neoplastic to sarcomatous neoplasia. With the mechanism of neoplastic transformation being incompletely understood, this study investigates if S. lupi parasite proteinaceous secretory/excretory products (ESPs) play a role in the neoplastic transformation. METHODS: To facilitate collection of ESPs, we maintained naturally harvested adult parasites in the laboratory under artificial conditions. Media in which the parasites were grown was subsequently evaluated for the presence of proteinaceous compounds using a mass spectroscopy library as well as for their ability to be mitogenic in primary murine fibroblastic cells. RESULTS: Chromatrography of the ethyl acetate extracted incubation media showed the presence of 9 protein compounds, of which three were identified as non-specific proteins isolated from Nematostella vectensis, Caenorhabditis brenneri and Sus scrofa, with the rest being unknown. Acetone, methanol, hexane and ethylacetate extracted culture media were unable to induce a mitogenic change in primary murine fibroblasts in comparison to the controls. CONCLUSION: While no mitogenic effect was evident, further studies are required to understand the role of worm excretory/secretory products on clastogenesis under chronic exposure. In addition, while not of primary importance for this study, the observed duration of parasite survival indicates that ex vivo studies on S. lupi are possible. For the latter we believe that the worm culture method can be further optimized if longer survival times are required.


Assuntos
Fibroblastos/parasitologia , Proteínas de Helminto/fisiologia , Mitógenos/fisiologia , Thelazioidea/patogenicidade , Animais , Transformação Celular Neoplásica , Células Cultivadas , Cães/parasitologia , Feminino , Proteínas de Helminto/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Thelazioidea/crescimento & desenvolvimento , Thelazioidea/isolamento & purificação
2.
Rev. neurol. (Ed. impr.) ; 64(12): 549-567, 16 jun., 2017. ilus, tab, graf
Artigo em Espanhol | IBECS | ID: ibc-164008

RESUMO

En el sistema nervioso central de los mamíferos, las células gliales superan diez veces en número a las neuronas. Su número permanente estacionario durante la edad adulta, controlado por la presencia simultánea de mitógenos gliales e inhibidores de esos mitógenos. El inhibidor más abundante, la neurostatina, es el gangliósido GD1b O-acetilado en el grupo 9 del ácido siálico más externo. La neurostatina y los oligosacáridos sintéticos inhiben la proliferación de astroblastos en cultivo primario (citostáticos) y de células de gliomas (citotóxicos), tanto de roedores como de humanos, en concentración nanomolar. A esas concentraciones, la neurostatina no tuvo efecto sobre células de linaje no glial ni sobre glía madura. La neurostatina y sus análogos mostraron actividad antimitótica directa sobre las células tumorales, interfiriendo con la progresión del ciclo celular en múltiples sitios, pero también actuaron indirectamente, haciendo visibles las células tumorales al sistema inmune del huésped y activando linfocitos CD4+ y CD8+. Análogos de neurostatina podrían generar nuevos fármacos antiinflamatorios, con múltiples acciones directas e indirectas contra el crecimiento de gliomas, una patología todavía sin tratamiento clínico satisfactorio. La neurostatina es producida por las neuronas, pero el contacto de éstas con astrocitos estimula notablemente su expresión. La acción de la neurostatina puede estar mediada por numerosas proteínas receptoras, incluyendo integrinas, siglecs y receptores Toll-like (AU)


Glial cells in the central nervous system of adult mammals outnumber neurons 10-fold. Their number remains stationary throughout adulthood, controlled by the concomitant presence of mitogens and mitogen inhibitors. The most abundant inhibitor, neurostatin, is ganglioside GD1b O-acetylated on hydroxyl 9 of its outermost sialic acid. Neurostatin inhibited the proliferation of primary microglia and astroblasts in culture (cytostatic) as well as both rodent and human glioma cells (cytotoxic) at nanomolar concentrations. At those concentrations neurostatin had no effect on non-glial lineage cells or differentiated glia. Neurostatin shows direct antimitotic activity on tumoral cells, interfering with multiple signals regulating cell cycle progression. But it also promotes indirectly total destruction of experimental rat brain glioma, presumably by making it visible to the host immune system and activating CD4+ and CD8+ lymphocytes. Neurostatin could be a new anti-inflammatory agent, with multiple convergent direct and indirect actions on glioma growth, a pathology without satisfactory clinical treatment. Neurostatin is produced by neurons but its expression is up-regulated by neuron-astrocyte contact. The action of neurostatin could be mediated by a number of receptor proteins, including integrins, Toll-like receptors and siglecs (AU)


Assuntos
Humanos , Glicolipídeos/farmacocinética , Neurônios/fisiologia , Células Ependimogliais/fisiologia , Neuroglia/fisiologia , Lesões Encefálicas Traumáticas/fisiopatologia , Divisão Celular/fisiologia , Mitógenos/fisiologia , Gangliosídeos/fisiologia , Receptores Toll-Like/fisiologia
3.
Hepatology ; 64(4): 1289-301, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27178107

RESUMO

UNLABELLED: Fibroblast growth factor 19 (FGF19) is an important postprandial enterokine which regulates liver metabolism and hepatocyte proliferation. However, the precise mechanism by which FGF19 regulates these cellular effects is poorly understood. Given that mechanistic target of rapamycin complex 1 (mTORC1) regulates numerous postprandial adaptations, we investigated the potential role of mTORC1 in FGF19 action. We found that FGF19 activated mTORC1 in HepG2 and HuH7 human hepatoma cells, differentiated 3T3-L1 adipocytes and mouse liver. FGF19 activates the mTORC1-p70S6K and extracellular signal-regulated kinase (Erk)-p90RSK pathways independently to regulate S6 in an additive manner in hepatoma cells, but it uses mTORC1 as the primary pathway to regulate S6 in 3T3-L1 adipocytes. Thus, mTORC1 is a novel mediator of FGF19 signaling, which can act in parallel with Erk or function as the primary pathway to regulate S6. The FGF19-induced mTORC1 pathway requires amino acids for efficient signaling; thus, involvement of mTORC1 confers amino acid sensitivity to FGF19 signaling. Although Akt and Erk are known to activate mTORC1, we found that FGF19 signals to mTORC1 through a third recently identified mTORC1 regulator, Ras-like (Ral) protein. Pharmacological or genetic inhibition of RalA or RalB abolished FGF19-induced mTORC1 activation, demonstrating that Ral proteins are required for FGF19 to activate mTORC1. FGF19 induced metabolic gene expression, fatty acid oxidation, cell growth, and proliferation in HepG2 cells; and these effects were abolished by mTORC1 inhibition, demonstrating an essential role of mTORC1 in FGF19 action. CONCLUSION: mTORC1 is a novel and essential mediator of FGF19 action on metabolic and mitogenic programs; thus, the involvement of mTORC1 in FGF19 signaling is an important factor to consider when targeting the pathway for cancer or diabetes therapy. (Hepatology 2016;64:1289-1301).


Assuntos
Carcinoma Hepatocelular/patologia , Fatores de Crescimento de Fibroblastos/fisiologia , Neoplasias Hepáticas/patologia , Complexos Multiproteicos/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Carcinoma Hepatocelular/metabolismo , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos BALB C , Mitógenos/fisiologia
4.
Mol Endocrinol ; 28(2): 197-207, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24422628

RESUMO

Insulin, an established mitogen that promotes breast cancer cell growth, has been implicated in the link between obesity and an increased risk of breast cancer. However, the current understanding of signaling pathways that mediate the mitogenic action of insulin remains incomplete. Here we provide the first evidence that insulin is capable of activating both sphingosine kinase (SphK) 1 and SphK 2, two isoenzymes that often exhibit opposing effects in the regulation of cell survival and growth. Insulin stimulates the phosphorylation of both SphK1 and SphK2 in a similar time- and dose-dependent manner. Interestingly, both isoenzymes are responsible equally for insulin-induced cell cycle progression and proliferation of MCF7 breast cancer cells, although SphK1 and SphK2 display different roles in mediating insulin-induced ERK1/2 and Akt activation. Moreover, the sphingosine 1-phosphate receptor 3, a key component of the SphK signaling system, is important for insulin-mediated mitogenic action in breast cancer cells. Furthermore, insulin receptor and type 1 IGF receptor (IGF1R) are responsible for the insulin-promoted mitogenic action on MCF7 breast cancer cells. Notably, IGF1R mediates insulin-stimulated phosphorylation of both SphK1 and SphK2, whereas insulin receptor is involved only in SphK1, but not SphK2, activation. Collectively the current study illustrates a new signaling system controlling the mitogenic action of insulin in breast cancer cells, suggesting a new strategy that pharmaceutically targets both isoenzymes of SphK for the management of breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Insulina/fisiologia , Mitógenos/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proliferação de Células , Feminino , Humanos , Isoenzimas/metabolismo , Células MCF-7 , Fosforilação , Processamento de Proteína Pós-Traducional , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato
5.
Mol Endocrinol ; 26(6): 1043-55, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496359

RESUMO

We previously demonstrated that long-term pretreatment of rat FRTL-5 thyroid cells with TSH or cAMP-generating reagents potentiated IGF-I-dependent DNA synthesis. Under these conditions, cAMP treatment increased tyrosine phosphorylation of a 125-kDa protein (p125) and its association with a p85 regulatory subunit of phosphatidylinositol 3-kinase (p85 PI3K), which were suggested to mediate potentiation of DNA synthesis. This study was undertaken to identify p125 and to elucidate its roles in potentiation of DNA synthesis induced by IGF-I. Matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) analysis revealed p125 to be a rat ortholog of human XB130, which we named PI3K-associated protein (PI3KAP). cAMP treatment elevated PI3KAP/XB130 mRNA and protein levels as well as tyrosine phosphorylation and interaction with p85 PI3K leading to increased PI3K activities associated with PI3KAP/XB130, supporting the role of PI3KAP/XB130 in DNA synthesis potentiation. Importantly, PI3KAP/XB130 knockdown attenuated cAMP-dependent potentiation of IGF-I-induced DNA synthesis. Furthermore, c-Src was associated with PI3KAP/XB130 and was activated in response to cAMP. Addition of Src family kinase inhibitors, PP1 or PP2, during cAMP treatment abolished tyrosine phosphorylation of PI3KAP/XB130 and its interaction with p85 PI3K. Finally, introduction of PI3KAP/XB130 into NIH3T3 fibroblasts lacking endogenous PI3KAP/XB130 enhanced IGF-I-induced DNA synthesis; however, a mutant Y72F incapable of binding to p85 PI3K did not show this response. Together, these data indicate that cAMP-dependent induction of PI3KAP/XB130, which is associated with PI3K, is required for enhancement of IGF mitogenic activities.


Assuntos
Proteínas de Transporte/metabolismo , AMP Cíclico/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Mitógenos/fisiologia , Glândula Tireoide/citologia , Animais , Proteínas de Transporte/genética , Ciclo Celular , Proliferação de Células , Replicação do DNA , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Análise de Sequência de DNA , Tireotropina/fisiologia , Quinases da Família src/metabolismo
6.
Life Sci ; 89(11-12): 364-70, 2011 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-21806999

RESUMO

AIM: To investigate the effects of shikonin on phorbol myristate acetate (PMA) plus cyclic adenosine monophosphate (cAMP)-induced T helper (T(H)) 2 cell cytokine production, and the underlying mechanism. MAIN METHODS: We used activated EL-4 murine T-lymphoma cells, which produce interleukin (IL)-4 and IL-5, but not interferon (IFN)-γ, as T(H)2 cell-like cells and treated them with PMA+cAMP to investigate the effects of shikonin on T(H)2 cytokines, transcriptional factors, and the related mitogen-activated protein kinase (MAPK)/nuclear factor (NF)-κB signaling pathway. KEY FINDINGS: The data show that shikonin inhibited the PMA+cAMP-induced mRNA and protein expression of IL-4 and IL-5 via the downregulation of GATA-binding protein-3 (GATA-3) and c-musculoaponeurotic fibrosarcoma (Maf) but not T-box expressed in T cells (T-bet). Moreover, shikonin suppressed the phosphorylation of p38, inhibitor of κB (IκB) kinase (IKK)-ß and IκB-α, and the subsequent IκB-α degradation induced by PMA+cAMP; however, the PMA+cAMP-induced phosphorylation of extracellular signal-related kinase (ERK), which resulted in minor inhibition and phosphorylation of c-Jun N-terminal kinase (JNK), seemed to be unaffected by shikonin treatment. SIGNIFICANCE: This study suggests that downregulation of GATA-3 and c-Maf via the suppression of p38, IKK-ß and IκB-α phosphorylation might contribute to the inhibitory effect of shikonin on mitogen-induced IL-4 and IL-5 production in EL-4T cells. Furthermore, shikonin is a potential drug for treating allergic diseases.


Assuntos
Antineoplásicos/farmacologia , Fator de Transcrição GATA3/metabolismo , Interleucina-4/biossíntese , Interleucina-5/biossíntese , Naftoquinonas/farmacologia , Proteínas Proto-Oncogênicas c-maf/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/toxicidade , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Citocinas/biossíntese , Citocinas/genética , Regulação para Baixo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/toxicidade , Quinase I-kappa B/antagonistas & inibidores , Proteínas I-kappa B/antagonistas & inibidores , Interleucina-4/genética , Interleucina-5/genética , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Mitógenos/fisiologia , Naftoquinonas/toxicidade , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
7.
Phytochemistry ; 72(16): 1947-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21798567

RESUMO

Cysteine proteinases from the Caricaceae belong to the C1 family of the CA clan and display papain-like structured, the archetype enzyme for this group of proteins. Carica candamarcensis, also named Vasconcellea cundinamarcensis, a member of Caricaceae family common to many areas in South America, contains cysteine proteinases with proteolytic activity five to eight-fold higher than those from latex of Carica papaya. The cysteine protease CMS2MS2 from C. candamarcensis latex has been shown to enhance proliferation of L929 fibroblast and to activate the extracellular signal-regulated protein kinase (ERK). In this study, the cDNA cloning, expression and evaluation of biological activity of a CMS2MS2-like protein from C. candamarcensis is reported. The 650 bp fragment was cloned in bacteria and the DNA sequence confirmed a cysteine-proteinase similar to CMS2MS2. The recombinant protein is 30 kDa, induces a mitogenic response, and enhances ERK1/2 phosphorylation, like the non-recombinant enzyme, but lacks either amidase or caseinolytic activity. The mitogenic activity of this protein and its lack of proteolytic activity underscore a potential for use in wound healing treatment.


Assuntos
Carica/enzimologia , Cisteína Proteases/fisiologia , Mitógenos/fisiologia , Proteínas de Plantas/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Clonagem Molecular , Cisteína Proteases/química , Cisteína Proteases/farmacologia , Camundongos , Mitógenos/química , Mitógenos/farmacologia , Dados de Sequência Molecular , Proteínas de Plantas/química , Proteínas de Plantas/farmacologia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Análise de Sequência de DNA , Análise de Sequência de Proteína , Cicatrização/efeitos dos fármacos
8.
PLoS One ; 6(1): e14565, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21283688

RESUMO

The development of different brain regions involves the coordinated control of proliferation and cell fate specification along and across the neuraxis. Here, we identify Plxdc2 as a novel regulator of these processes, using in ovo electroporation and in vitro cultures of mammalian cells. Plxdc2 is a type I transmembrane protein with some homology to nidogen and to plexins. It is expressed in a highly discrete and dynamic pattern in the developing nervous system, with prominent expression in various patterning centres. In the chick neural tube, where Plxdc2 expression parallels that seen in the mouse, misexpression of Plxdc2 increases proliferation and alters patterns of neurogenesis, resulting in neural tube thickening at early stages. Expression of the Plxdc2 extracellular domain alone, which can be cleaved and shed in vivo, is sufficient for this activity, demonstrating a cell non-autonomous function. Induction of proliferation is also observed in cultured embryonic neuroepithelial cells (ENCs) derived from E9.5 mouse neural tube, which express a Plxdc2-binding activity. These experiments uncover a direct molecular activity of Plxdc2 in the control of proliferation, of relevance in understanding the role of this protein in various cancers, where its expression has been shown to be altered. They also implicate Plxdc2 as a novel component of the network of signalling molecules known to coordinate proliferation and differentiation in the developing nervous system.


Assuntos
Proteínas Aviárias/fisiologia , Mitógenos/fisiologia , Células-Tronco Neurais/citologia , Neurogênese , Receptores de Superfície Celular/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Embrião de Galinha , Tubo Neural
9.
Am J Physiol Cell Physiol ; 300(3): C517-25, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148410

RESUMO

Arterial smooth muscle cells enter the cell cycle and proliferate in conditions of disease and injury, leading to adverse vessel remodeling. In the pulmonary vasculature, diverse stimuli cause proliferation of pulmonary artery smooth muscle cells (PASMCs), pulmonary artery remodeling, and the clinical condition of pulmonary hypertension associated with significant health consequences. PASMC proliferation requires extracellular Ca(2+) influx that is intimately linked with intracellular Ca(2+) homeostasis. Among the primary sources of Ca(2+) influx in PASMCs is the low-voltage-activated family of T-type Ca(2+) channels; however, up to now, mechanisms for the action of T-type channels in vascular smooth muscle cell proliferation have not been addressed. The Ca(v)3.1 T-type Ca(2+) channel mRNA is upregulated in cultured PASMCs stimulated to proliferate with insulin-like growth factor-I (IGF-I), and this upregulation depends on phosphatidylinositol 3-kinase/Akt signaling. Multiple stimuli that trigger an acute rise in intracellular Ca(2+) in PASMCs, including IGF-I, also require the expression of Ca(v)3.1 Ca(2+) channels for their action. IGF-I also led to cell cycle initiation and proliferation of PASMCs, and, when expression of the Ca(v)3.1 Ca(2+) channel was knocked down by RNA interference, so were the expression and activation of cyclin D, which are necessary steps for cell cycle progression. These results confirm the importance of T-type Ca(2+) channels in proper progression of the cell cycle in PASMCs stimulated to proliferate by IGF-I and suggest that Ca(2+) entry through Ca(v)3.1 T-type channels in particular interacts with Ca(2+)-dependent steps of the mitogenic signaling cascade as a central component of vascular remodeling in disease.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Animais , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/fisiologia , Membrana Celular/metabolismo , Proliferação de Células , Células Cultivadas , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertrofia , Mitógenos/genética , Mitógenos/fisiologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/patologia , Ratos , Transdução de Sinais/genética
10.
Cell Res ; 20(10): 1148-57, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20628369

RESUMO

Animal cells require extrinsic cues for growth, proliferation and survival. The propagation of Drosophila imaginal disc cells in vitro, for example, requires the supplementation of fly extract, the composition of which remains largely undefined. Here I report the biochemical purification of iron-loaded ferritin as an active ingredient of fly extract that is required for promoting the growth of clone 8 imaginal disc cells. Consistent with an essential role for iron-loaded ferritin in cultured cells, overexpression of ferritin or addition of iron in a nutrient-poor diet increases animal viability and body weight, promotes cell proliferation, and shortens the duration of postembryonic development. Conversely, overexpression of dominant-negative ferritin or addition of iron chelator causes the opposite effects. Ferritin mutant flies arrest development at the first-instar larval stage with a severe starvation phenotype reminiscent of that seen in starved larvae. I conclude that iron-loaded ferritin acts as an essential mitogen for cell proliferation and postembryonic development in Drosophila by maintaining iron homeostasis and antagonizing starvation response.


Assuntos
Drosophila/crescimento & desenvolvimento , Ferritinas/fisiologia , Ferro/química , Mitógenos/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Drosophila/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Ferritinas/genética , Ferritinas/farmacologia , Quelantes de Ferro/farmacologia , Larva/efeitos dos fármacos , Larva/metabolismo , Mitógenos/farmacologia , Mutação
11.
Genes Dev ; 24(13): 1377-88, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20551164

RESUMO

Loss of G1/S control is a hallmark of cancer, and is often caused by inactivation of the retinoblastoma pathway. However, mouse embryonic fibroblasts lacking the retinoblastoma genes RB1, p107, and p130 (TKO MEFs) are still subject to cell cycle control: Upon mitogen deprivation, they enter and complete S phase, but then firmly arrest in G2. We now show that G2-arrested TKO MEFs have accumulated DNA damage. Upon mitogen readdition, cells resume proliferation, although only part of the damage is repaired. As a result, mitotic cells show chromatid breaks and chromatid cohesion defects. These aberrations lead to aneuploidy in the descendent cell population. Thus, our results demonstrate that unfavorable growth conditions can cause genomic instability in cells lacking G1/S control. This mechanism may allow premalignant tumor cells to acquire additional genetic alterations that promote tumorigenesis.


Assuntos
Instabilidade Genômica , Mitógenos/fisiologia , Proteína do Retinoblastoma , Proteína p107 Retinoblastoma-Like , Proteína p130 Retinoblastoma-Like , Transdução de Sinais/fisiologia , Aneuploidia , Animais , Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Centrômero , Quebras de DNA de Cadeia Dupla , Variações do Número de Cópias de DNA , Fibroblastos/citologia , Camundongos , Mitógenos/farmacologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like/deficiência , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/deficiência , Proteína p130 Retinoblastoma-Like/genética , Proteína p130 Retinoblastoma-Like/metabolismo
12.
Int J Biochem Cell Biol ; 42(5): 749-54, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20083221

RESUMO

Advanced glycation end products (AGEs) are formed by the non-enzymatic glycation of proteins by reducing carbohydrates or alpha-oxo-aldehydes such as glyoxal and methylglyoxal and further rearrangements, eliminations and oxidations. AGE-modifications alter peptide structure, function and stability and accumulate under several pathophysiological conditions such as diabetes and are considered a biomarker of ageing. PDGF is a major regulator of wound healing, which is impaired in hyperglycaemia and ageing. We analyzed whether glycated PDGF has impaired activity in cell culture models and occurs in human subjects. PDGF was AGE-modified by the alpha-oxo-aldehydes glyoxal and methylglyoxal, which was shown by Western-blotting using alpha-carboxymethyllysine (CML) or alpha-arginine-pyrimidine (Arg-Pyr) antibodies. In mouse AKR-2B fibroblasts, this AGE-modified PDGF exhibited reduced signalling to AKT and ERK resulting in decreased cell proliferation. In the human osteosarcoma cell line 143B, PDGF signalling towards the AKT-kinase was decreased when using modified PDGF-AA, -AB, and -BB whereas the constitutive active ERK was not affected. Secreted proteins from collagen-activated platelets from diabetic subjects contained more CML-modified proteins compared to healthy controls. PDGF protein as a platelet protein coprecipitated in immunoprecipitation experiments with alpha-CML-antiserum. In summary, our data suggest that AGE-modification of PDGF contributes to reduced wound healing in diabetic patients.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Mitógenos/fisiologia , Ativação Plaquetária , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Animais , Becaplermina , Plaquetas/metabolismo , Linhagem Celular , Proliferação de Células , Diabetes Mellitus/sangue , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Produtos Finais de Glicação Avançada/sangue , Glicosilação , Glioxal/química , Humanos , Hiperglicemia/sangue , Masculino , Camundongos , Fosforilação , Projetos Piloto , Multimerização Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis , Aldeído Pirúvico/química , Proteínas Recombinantes/química , Cicatrização
13.
J Cell Biol ; 182(5): 837-43, 2008 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-18762583

RESUMO

Directed cell migration requires the orientation of the Golgi and centrosome toward the leading edge. We show that stimulation of interphase cells with the mitogens epidermal growth factor or lysophosphatidic acid activates the extracellular signal-regulated kinase (ERK), which phosphorylates the Golgi structural protein GRASP65 at serine 277. Expression of a GRASP65 Ser277 to alanine mutant or a GRASP65 1-201 truncation mutant, neither of which can be phosphorylated by ERK, prevents Golgi orientation to the leading edge in a wound assay. We show that phosphorylation of GRASP65 with recombinant ERK leads to the loss of GRASP65 oligomerization and causes Golgi cisternal unstacking. Furthermore, preventing Golgi polarization by expressing mutated GRASP65 inhibits centrosome orientation, which is rescued upon disassembly of the Golgi structure by brefeldin A. We conclude that Golgi remodeling, mediated by phosphorylation of GRASP65 by ERK, is critical for the establishment of cell polarity in migrating cells.


Assuntos
Polaridade Celular , Centrossomo/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Animais , Brefeldina A/farmacologia , Movimento Celular/fisiologia , Ativação Enzimática/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Complexo de Golgi/efeitos dos fármacos , Proteínas da Matriz do Complexo de Golgi , Interfase , Lisofosfolipídeos/farmacologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mitógenos/fisiologia , Fosforilação , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/farmacologia , Ratos
14.
J Immunol ; 181(7): 5054-61, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802109

RESUMO

The species Staphylococcus aureus harbors 19 superantigen gene loci, six of which are located in the enterotoxin gene cluster (egc). Although these egc superantigens are far more prevalent in clinical S. aureus isolates than non-egc superantigens, they are not a prominent cause of toxic shock. Moreover, neutralizing Abs against egc superantigens are very rare, even among carriers of egc-positive S. aureus strains. In search of an explanation, we have tested two non-exclusive hypotheses: 1) egc and non-egc superantigens have unique intrinsic properties and drive the immune system into different directions and 2) egc and non-egc superantigens are released by S. aureus under different conditions, which shape the immune response. A comparison of three egc (SEI, SElM, and SElO) and three non-egc superantigens (SEB, SElQ, and toxic shock syndrome toxin-1) revealed that both induced proliferation of human PBMC with comparable potency and elicited similar Th1/Th2-cytokine signatures. This was supported by gene expression analysis of PBMC stimulated with one representative superantigen from each group (SEI and SEB). They induced very similar transcriptional changes, especially of inflammation-associated gene networks, corresponding to a very strong Th1- and Th17-dominated immune response. In contrast, the regulation of superantigen release differed markedly between both superantigen groups. Egc-encoded proteins were secreted by S. aureus during exponential growth, while non-egc superantigens were released in the stationary phase. We conclude that the distinct biological behavior of egc and non-egc superantigens is not due to their intrinsic properties, which are very similar, but caused by their differential release by S. aureus.


Assuntos
Toxinas Bacterianas/genética , Enterotoxinas/genética , Família Multigênica/imunologia , Staphylococcus aureus/imunologia , Superantígenos/genética , Proliferação de Células , Citocinas/biossíntese , Enterotoxinas/metabolismo , Enterotoxinas/fisiologia , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/imunologia , Humanos , Mediadores da Inflamação/fisiologia , Mitógenos/genética , Mitógenos/metabolismo , Mitógenos/fisiologia , Staphylococcus aureus/genética , Staphylococcus aureus/isolamento & purificação , Superantígenos/metabolismo , Superantígenos/fisiologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/microbiologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/microbiologia
15.
Mol Pharmacol ; 73(2): 566-74, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17993511

RESUMO

Excessive smooth muscle growth occurs within the context of inflammation associated with certain vascular and airway diseases. The inflammatory cytokines interleukin (IL)-1beta and tumor necrosis factor-alpha (TNF-alpha) have been shown previously to inhibit mitogen-stimulated smooth muscle growth through a mechanism presumed to be dependent on the induction of cyclooxygenase-2, prostaglandins, and activation of the cAMP-dependent protein kinase (PKA). Using both molecular and pharmacological strategies, we demonstrate that the mitogenic effects of IL-1beta and TNF-alpha on cultured human airway smooth muscle (ASM) cells are tightly regulated by PKA activity. Suppression of induced PKA activity by either corticosteroids or cyclooxygenase inhibitors converts the cytokines from inhibitors to enhancers of mitogen-stimulated ASM growth, and biological variability in the capacity to activate PKA influences the modulatory effect of cytokines. Promitogenic effects of IL-1beta are associated with delayed increases in p42/p44 and phosphoinositide-3 kinase activities, suggesting a role for induced autocrine factors. These findings suggest a mechanism by which mainstream therapies such as corticosteroids or cyclooxygenase inhibitors could fail to address or exacerbate the pathogenic smooth muscle growth that occurs in obstructive airway and cardiovascular diseases.


Assuntos
Corticosteroides/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Citocinas/fisiologia , Mitógenos/fisiologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Citocinas/genética , Humanos , Interleucina-1beta/genética , Interleucina-1beta/fisiologia , Mitógenos/genética
17.
J Rheumatol ; 34(1): 20-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17216675

RESUMO

OBJECTIVE: A3 adenosine receptor (A3AR) upregulation has been found in cells of synovial tissue and in peripheral blood mononuclear cells (PBMC) of rats with adjuvant-induced arthritis. We investigated A3AR levels in PBMC of patients with rheumatoid arthritis (RA) and in mitogen-activated PBMC from healthy subjects. We examined the role of nuclear factor-kappaB (NF-kappaB), a transcription factor present in the A3AR promoter, in mediating receptor upregulation. METHODS: A3AR and NF-kappaB protein levels were evaluated in PBMC of RA patients (n = 23) and healthy subjects by Western blot. A3AR and NF-kappaB levels were also analyzed in phytohemagglutinin (PHA) and lipopolysaccharide (LPS)-stimulated PBMC in the presence and absence of antibodies against interleukin 2 (IL-2) and tumor necrosis factor-alpha (TNF-alpha). Reverse transcription-polymerase chain reaction was performed in PHA-stimulated PBMC of healthy subjects to determine A3AR expression. RESULTS: A3AR was overexpressed in PBMC of RA patients compared to healthy subjects and was directly correlated to an increase in NF-kappaB. Similar findings were observed in PHA and LPS-stimulated PBMC from healthy subjects. Antibodies against IL-2 or TNF-alpha prevented the increase in A3AR and NF-kappaB expression. CONCLUSION: Overexpression of A3AR was found in PBMC of RA patients. Receptor upregulation was induced by inflammatory cytokines controlling the expression of the A3AR transcription factor NF-kappaB.


Assuntos
Artrite Reumatoide/sangue , Leucócitos Mononucleares/metabolismo , Receptor A3 de Adenosina/sangue , Adenosina/análogos & derivados , Adenosina/farmacologia , Artrite Reumatoide/metabolismo , Regulação da Expressão Gênica , Humanos , Interleucina-2/antagonistas & inibidores , Leucócitos Mononucleares/patologia , Pessoa de Meia-Idade , Mitógenos/fisiologia , NF-kappa B/fisiologia , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Receptor A3 de Adenosina/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores
18.
Glia ; 55(5): 537-45, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17236210

RESUMO

Mitogenic growth factors play an important role in the initial stages of oligodendrocyte development, but their roles in the process of myelination itself remain less well defined. In order to study directly the effects of different growth factors on myelination, we used a purified in vitro co-culture system with dorsal root ganglion neurons and oligodendrocytes. Extensive myelination had occurred in these cultures 14 days after oligodendrocyte precursors (OPCs) were added, with the relationship between neurite density and the percentage of oligodendrocytes forming myelin sheaths providing a robust and straightforward means of quantifying myelination. Addition of soluble neuregulin (Nrg1), a mitogen for oligodendroglial cells that also provides an axonal signal implicated in oligodendrocyte survival, increased myelination. Conversely, the OPC mitogens FGF-2 and PDGF inhibited myelination. The inhibitory effect of these mitogens was reversible, as inhibition of PDGF allowed myelination to proceed. Taken together, these data indicate that different mitogenic growth factors can regulate myelination by oligodendrocytes in addition to their well-described effects on earlier stages of oligodendroglial development. Moreover, the results highlight important differences between the growth factors.


Assuntos
Bainha de Mielina/metabolismo , Fatores de Crescimento Neural/fisiologia , Neuregulina-1/fisiologia , Neurônios/metabolismo , Oligodendroglia/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Fator 2 de Crescimento de Fibroblastos/fisiologia , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Mitógenos/fisiologia , Neuritos/metabolismo , Neuritos/ultraestrutura , Neurônios/ultraestrutura , Oligodendroglia/ultraestrutura , Fator de Crescimento Derivado de Plaquetas/fisiologia , Ratos , Células-Tronco/citologia
19.
Nat Rev Neurosci ; 7(10): 772-83, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16988653

RESUMO

Sonic hedgehog has received an enormous amount of attention since its role as a morphogen that directs ventral patterning in the spinal cord was discovered a decade ago. Since that time, a bewildering array of information has been generated concerning both the components of the hedgehog signalling pathway and the remarkable number of contexts in which it functions. Nowhere is this more evident than in the nervous system, where hedgehog signalling has been implicated in events as disparate as axonal guidance and stem cell maintenance. Here we review our present knowledge of the hedgehog signalling pathway and speculate about areas in which further insights into this versatile pathway might be forthcoming.


Assuntos
Mitógenos/fisiologia , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Transativadores/fisiologia , Animais , Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog , Modelos Biológicos
20.
J Virol ; 80(21): 10554-64, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16920820

RESUMO

The hepatitis B virus (HBV) X protein (pX) is implicated in hepatocarcinogenesis by an unknown mechanism. pX variants encoded by HBV genomes found integrated in genomic DNA from liver tumors of patients with hepatocellular carcinoma (HCC) generally lack amino acids 134 to 154. Since deregulation of mitogenic pathways is linked to oncogenic transformation, herein we define the pX region required for mitogenic pathway activation. A series of pX deletions was used to construct tetracycline-regulated pX-expressing cell lines. The activation of the mitogenic pathways by these pX deletions expressed in the constructed cell lines was measured by transient transreporter assays, effects on endogenous cyclin A expression, and apoptosis. Conditional expression of pX51-140 in AML12 clone 4 cell line activates the mitogenic pathways, induces endogenous cyclin A expression, and sensitizes cells to apoptosis, similar to wild-type (WT) pX. By contrast, pX1-115 is inactive, supporting the idea that amino acids 116 to 140 are required for mitogenic pathway activation. Moreover, this pX deletion analysis demonstrates that WT pX function is modulated by two regions spanning amino acids 1 to 78 and 141 to 154. The N-terminal X1-78, expressed via a retroviral vector in WT pX-expressing 4pX-1 cells, coimmunoprecipitates with WT pX, indicating this pX region participates in protein-protein interactions leading to pX oligomerization. Interestingly, pX1-78 interferes with WT pX in mediating mitogenic pathway activation, endogenous gene expression, and apoptosis. The C-terminal pX region spanning amino acids 141 to 154 decreases pX stability, determined by pulse-chase studies of WT pX and pX1-140, suggesting that increased stability of naturally occurring pX variants lacking amino acids 134 to 154 may play a role in HCC development.


Assuntos
Vírus da Hepatite B/fisiologia , Transativadores/química , Transativadores/fisiologia , Animais , Apoptose , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/virologia , Linhagem Celular , Ciclina A/genética , Ciclina A/metabolismo , Expressão Gênica , Genes Virais , Vírus da Hepatite B/genética , Vírus da Hepatite B/patogenicidade , Humanos , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/virologia , Camundongos , Mitógenos/química , Mitógenos/genética , Mitógenos/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transdução de Sinais , Tetraciclina/farmacologia , Transativadores/genética , Transfecção , Proteínas Virais Reguladoras e Acessórias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...