Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Radiat Res ; 186(2): 162-74, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27388041

RESUMO

There is increasing evidence that radiation-induced damage to endothelial cells and loss of endothelial function may contribute to both acute radiation syndromes and long-term effects of whole-body nuclear irradiation. Therefore, several drugs are being developed to mitigate the effects of nuclear radiation, most of these drugs will target and protect or regenerate leukocytes and platelets. Our laboratory has demonstrated that TP508, a 23-amino acid thrombin peptide, activates endothelial cells and stem cells to revascularize and regenerate tissues. We now show that TP508 can mitigate radiation-induced damage to endothelial cells in vitro and in vivo. Our in vitro results demonstrate that human endothelial cells irradiation attenuates nitric oxide (NO) signaling, disrupts tube formation and induces DNA double-strand breaks (DSB). TP508 treatment reverses radiation effects on NO signaling, restores tube formation and accelerates the repair of radiation-induced DSB. The radiation-mitigating effects of TP508 on endothelial cells were also seen in CD-1 mice where systemic injection of TP508 stimulated endothelial cell sprouting from aortic explants after 8 Gy irradiation. Systemic doses of TP508 that mitigated radiation-induced endothelial cell damage, also significantly increased survival of CD-1 mice when injected 24 h after 8.5 Gy exposure. These data suggest that increased survival observed with TP508 treatment may be due to its effects on vascular and microvascular endothelial cells. Our study supports the usage of a regenerative drug such as TP508 to activate endothelial cells as a countermeasure for mitigating the effects of nuclear radiation.


Assuntos
Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/efeitos da radiação , Fragmentos de Peptídeos/farmacologia , Trombina/farmacologia , Sequência de Aminoácidos , Animais , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Relação Dose-Resposta à Radiação , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Masculino , Camundongos , Óxido Nítrico/biossíntese , Análise de Sobrevida
2.
J Vasc Res ; 50(3): 186-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23594718

RESUMO

Revascularization of chronic wounds and ischemic tissue is attenuated by endothelial dysfunction and the inability of angiogenic factors to stimulate angiogenesis. We recently showed that TP508, a nonproteolytic thrombin peptide, increases perfusion and NO-dependent vasodilation in hearts with chronic ischemia and stimulates NO production by endothelial cells. In this study, we investigated systemic in vivo effects of TP508 on VEGF-stimulated angiogenesis in vitro using aortic explants in normoxic and hypoxic conditions. Mice were injected with saline or TP508 and 24 h later aortas were removed and cultured to quantify endothelial sprouting. TP508 injection increased endothelial sprouting and potentiated the in vitro response to VEGF. Exposure of control explants to hypoxia inhibited basal and VEGF-stimulated endothelial cell sprouting. This effect of hypoxia was significantly prevented by TP508 injection. Thus, TP508 systemic administration increases responsiveness of aortic endothelial cells to VEGF and diminishes the effect of chronic hypoxia on endothelial cell sprouting. Studies using human endothelial cells in culture suggest that protective effects of TP508 during hypoxia may involve stimulation of endothelial cell NO production. These data suggest potential clinical benefit of using a combination of systemic TP508 and local VEGF as a therapy for revascularization of ischemic tissue.


Assuntos
Indutores da Angiogênese , Hipóxia Celular/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Trombina/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/farmacologia , Sequência de Aminoácidos , Animais , Aorta Torácica/citologia , Aorta Torácica/fisiologia , Sinergismo Farmacológico , Quimioterapia Combinada , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Trombina/química , Técnicas de Cultura de Tecidos
3.
J Vasc Res ; 47(3): 203-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19893317

RESUMO

TP508, a 23-amino-acid peptide representing a portion of human thrombin, promotes tissue revascularization and repair. The molecular mechanisms of TP508 action, however, remain unclear. Nitric oxide (NO) plays a crucial role in regulation of angiogenesis and wound healing. We, therefore, investigated TP508 effects on NO production in human endothelial cells. TP508 stimulated a rapid, dose-dependent, 2- to 4-fold increase in NO production. TP508 induced NO release as early as 5 min. Continued exposure to TP508 for 1-24 h increased NO concentrations over controls by 100.5 +/- 9.6 and 463.3 +/- 24.2 nM, respectively. These levels of NO release were similar to those produced in response to vascular endothelial growth factor (VEGF). TP508- and VEGF-induced NO production was decreased by inhibitors of PI-3K (LY294002) and Src (PP2). TP508 stimulated early transient phosphorylation of Src and Akt. In contrast to VEGF, TP508 stimulation of NO release was inhibited by PKC inhibitor (Go6976) and was independent of intracellular calcium mobilization. These results demonstrate that TP508 and VEGF stimulate NO production to similar levels but through distinct pathways. This study provides new insights into the initial molecular mechanisms by which TP508 may stimulate diverse cellular effects leading to tissue revascularization and wound healing.


Assuntos
Células Endoteliais/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fragmentos de Peptídeos/farmacologia , Trombina/farmacologia , Animais , Cálcio/metabolismo , Bovinos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
4.
Am J Physiol Cell Physiol ; 296(5): C1162-70, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244479

RESUMO

Coronary artery disease results in progressive vascular stenosis associated with chronic myocardial ischemia. Vascular endothelial growth factor (VEGF) stimulates endothelial cell angiogenic responses to revascularize ischemic tissues; however, the effect of chronic hypoxia on the responsiveness of endothelial cells to VEGF remains unclear. We, therefore, investigated whether hypoxia alters VEGF-stimulated signaling and angiogenic responses in primary human coronary artery endothelial (HCAE) cells. Exposure of HCAE cells to hypoxia (1% O(2)) for 24 h decreased VEGF-stimulated endothelial cell migration ( approximately 82%), proliferation ( approximately 30%), and tube formation. Hypoxia attenuated VEGF-stimulated activation of endothelial nitric oxide (NO) synthase (eNOS) ( approximately 72%) and reduced NO production in VEGF-stimulated cells from 237 +/- 38.8 to 61.3 +/- 28.4 nmol/l. Moreover, hypoxia also decreased the ratio of phosphorylated eNOS to total eNOS in VEGF-stimulated cells by approximately 50%. This effect was not observed in thrombin-stimulated cells, suggesting that hypoxia specifically inhibited VEGF signaling upstream of eNOS phosphorylation. VEGF-induced activation of Akt, ERK1/2, p38, p70S6 kinases, and S6 ribosomal protein was also attenuated in hypoxic cells. Moreover, VEGF-stimulated phosphorylation of VEGF receptor-2 (KDR) at Y996 and Y1175 was decreased by hypoxia. This decrease correlated with a 70 +/- 12% decrease in KDR protein expression. Analysis of mRNA from these cells showed that hypoxia reduced steady-state levels of KDR mRNA by 52 +/- 16% and decreased mRNA stability relative to normoxic cells. Our findings demonstrate that chronic hypoxia attenuates VEGF-stimulated signaling in HCAE cells by specific downregulation of KDR expression. These data provide a novel explanation for the impaired angiogenic responses to VEGF in endothelial cells exposed to chronic hypoxia.


Assuntos
Células Endoteliais/metabolismo , Hipóxia/metabolismo , Isquemia Miocárdica/metabolismo , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Capilares/citologia , Capilares/metabolismo , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Doença Crônica , Vasos Coronários/citologia , Regulação para Baixo/fisiologia , Células Endoteliais/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/fisiologia , Humanos , Hipóxia/patologia , Isquemia Miocárdica/patologia , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Cardiovasc Pharmacol Ther ; 13(3): 214-25, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18757834

RESUMO

Endothelial dysfunction (ED) is characterized by impaired nitric oxide (NO) signaling, decreased NO-dependent vasodilatation, increased vascular inflammation, and diminished response to angiogenic factors. TP508 (Chrysalin), an angiogenic tissue repair peptide, was tested for potential effects on myocardial revascularization and ED using a porcine model of chronic myocardial ischemia. TP508 increased perfusion in ischemic regions up to16-fold (P < .02) and doubled myocardial wall thickening (P < .02) relative to placebo controls. Ischemic arterioles exhibited impaired NO-mediated vasodilation and diminished NO production. TP508 reversed ischemic effects, increasing NO-mediated vasodilation (P < .05), endothelial nitric oxide synthase (eNOS) expression, and NO production. In human endothelial cells, TP508 stimulated eNOS activation (1.84 +/- 0.2-fold; P < .02), increased NO production (85 +/- 18%; P < .02), and prevented hypoxia-induced eNOS downregulation (P < .01). Thus, TP508 reverses ED both in porcine ischemic hearts and cultured human endothelial cells. These results suggest potential therapeutic benefit of TP508 in myocardial revascularization and treatment of ED-related diseases.


Assuntos
Indutores da Angiogênese/farmacologia , Circulação Coronária/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Isquemia Miocárdica/tratamento farmacológico , Revascularização Miocárdica/métodos , Fragmentos de Peptídeos/farmacologia , Animais , Hipóxia Celular , Células Cultivadas , Doença Crônica , Angiografia Coronária , Vasos Coronários/metabolismo , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ecocardiografia sob Estresse , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Humanos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Suínos , Porco Miniatura , Trombina , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
6.
Expert Opin Pharmacother ; 9(15): 2717-26, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18803458

RESUMO

Rusalatide acetate (Chrysalin) is an investigational drug being evaluated for treatment of chronic wounds and fractures. Rusalatide acetate interacts with cell surface receptors to stimulate a cascade of cellular and molecular wound healing events, including activation of nitric oxide signaling. Rusalatide acetate significantly accelerated healing of diabetic foot ulcers and distal radius fractures in Phase I/II clinical trials. Subsequently, in one of the largest Phase III fracture studies to date, rusalatide acetate showed significant acceleration of distal radius fracture healing radiographically but failed to meet its primary clinical endpoint - time to removal of immobilization - within the intent-to-treat population. Subset analysis showed that rusalatide acetate met this primary clinical endpoint and significantly accelerated radiographic healing in osteopenic women. Rusalatide acetate may therefore show its greatest efficacy in healing-impaired patients.


Assuntos
Pé Diabético/tratamento farmacológico , Consolidação da Fratura/efeitos dos fármacos , Fragmentos de Peptídeos/uso terapêutico , Trombina/uso terapêutico , Sequência de Aminoácidos , Ensaios Clínicos como Assunto , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacocinética , Fragmentos de Peptídeos/farmacologia , Trombina/efeitos adversos , Trombina/química , Trombina/farmacocinética , Trombina/farmacologia
7.
J Immunol ; 180(6): 4182-90, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322230

RESUMO

Reversal of eosinophilic inflammation has been an elusive therapeutic goal in the management of asthma pathogenesis. In this regard, GM-CSF is a primary candidate cytokine regulating eosinophil activation and survival in the lung; however, its molecular mechanism of propagation and maintenance of stimulated eosinophil activation is not well understood. In this study, we elucidate those late interactions occurring between the GM-CSF receptor and activated eosinophil signaling molecules. Using coimmunoprecipitation with GM-CSF-stimulated eosinophils, we have identified that the GM-CSF receptor beta-chain (GMRbeta) interacted with ICAM-1 and Shp2 phosphatase, as well as Slp76 and ADAP adaptor proteins. Separate experiments using affinity binding with a tyrosine-phosphorylated peptide containing an ITIM (ICAM-1 residues 480-488) showed binding to Shp2 phosphatase and GMRbeta. However, the interaction of GMRbeta with the phosphorylated ICAM-1-derived peptide was observed only with stimulated eosinophil lysates, suggesting that the interaction of GMRbeta with ICAM-1 required phosphorylated Shp2 and/or phosphorylated GMRbeta. Importantly, we found that inhibition of ICAM-1 in activated eosinophils blocked GM-CSF-induced expression of c-fos, c-myc, IL-8, and TNF-alpha. Moreover, inhibition of ICAM-1 expression with either antisense oligonucleotide or an ICAM-1-blocking Ab effectively inhibited ERK activation and eosinophil survival. We concluded that the interaction between ICAM-1 and the GM-CSF receptor was essential for GM-CSF-induced eosinophil activation and survival. Taken together, these results provide novel mechanistic insights defining the interaction between ICAM-1 and the GM-CSF receptor and highlight the importance of targeting ICAM-1 and GM-CSF/IL-5/IL-3 receptor systems as a therapeutic strategy to counter eosinophilia in asthma.


Assuntos
Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Eosinófilos/citologia , Eosinófilos/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Receptor Cross-Talk , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transdução de Sinais , Sobrevivência Celular/imunologia , Células Cultivadas , Subunidade beta Comum dos Receptores de Citocinas/fisiologia , Eosinófilos/enzimologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Humanos , Molécula 1 de Adesão Intercelular/fisiologia , Peptídeos/metabolismo , Peptídeos/fisiologia , Ligação Proteica/imunologia , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Receptor Cross-Talk/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/fisiologia , Transdução de Sinais/imunologia
8.
J Biol Chem ; 279(39): 40400-4, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15292208

RESUMO

Src activity is elevated in a majority of colonic and pancreatic cancers and is associated with late stage aggressive cancers. However, the mechanisms leading to its increased activity remain largely undefined. Agonist binding to the cholecystokinin-2 (CCK2)/gastrin receptor (CCK2R), a G-protein-coupled receptor, increases Src activity in a variety of normal and neoplastic cell lines. Recently, we and others (Hellmich, M. R., Rui, X. L., Hellmich, H. L., Fleming, R. Y., Evers, B. M., and Townsend, C. M., Jr. (2000) J. Biol. Chem. 275, 32122-32128; Ding, W. Q., Kuntz, S. M., and Miller, L. J. (2002) Cancer Res. 62, 947-952; Smith, J. P., Verderame, M. F., McLaughlin, P., Martenis, M., Ballard, E., and Zagon, I. S. (2002) Int. J. Mol. Med. 10, 689-694) have identified a splice variant of CCK2R, called CCK2i4svR, that is expressed in human colorectal and pancreatic cancers but not by cells of the adjacent nonmalignant tissue. Compared with CCK2R, CCK2i4svR contains an additional 69 amino acids within its third intracellular loop (3il) domain. Because CCK2i4svR is the only splice variant expressed in some human colon and pancreatic cancers, we questioned whether CCK2i4svR could regulate Src activity. Stably transfected HEK293 cells were used because, unlike many cancer-derived cells, they have a low level of basal Src activity. We report that, in contrast to CCK2R, CCK2i4svR activates Src kinase in the absence of agonist stimulation. In vitro kinase assay of immunoprecipitated receptor protein showed a 6-8-fold increase in Src kinase activity associated with CCK2i4svR compared with CCK2R. Expression of the 3il domain of the CCK2i4svR alone was sufficient to partially activate Src kinase. Together, these data support the hypothesis that the increased Src activity observed in some pancreatic and colorectal cancers is due, in part, to the co-expression of CCK2i4svR.


Assuntos
Receptor de Colecistocinina B/genética , Quinases da Família src/metabolismo , Processamento Alternativo , Aminoácidos/química , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Ativação Enzimática , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Receptor de Colecistocinina B/metabolismo , Fatores de Tempo , Transfecção
9.
J Biol Chem ; 279(3): 1853-60, 2004 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-14602717

RESUMO

Small differences in amplitude, duration, and temporal patterns of change in the concentration of free intracellular Ca2+ ([Ca2+](i)) can profoundly affect cell physiology, altering programs of gene expression, cell proliferation, secretory activity, and cell survival. We report a novel mechanism for amplitude modulation of [Ca2+](i) that involves mitogen-activated protein kinase (MAPK). We show that epidermal growth factor (EGF) potentiates gastrin-(1-17) (G17)-stimulated Ca2+ release from intracellular Ca2+ stores through a MAPK-dependent pathway. G17 activation of the cholecystokinin/gastrin receptor (CCK(2)R), a G protein-coupled receptor, stimulates release of Ca2+ from inositol 1,4,5-triphosphate-sensitive Ca2+ stores. Pretreating rat intestinal epithelial cells expressing CCK(2)R with EGF increased the level of G17-stimulated Ca2+ release from intracellular stores. The stimulatory effect of EGF on CCK(2)R-mediated Ca2+ release requires activation of the MAPK kinase (MEK)1,2/extracellular signal-regulated kinase (ERK)1,2 pathway. Inhibition of the MEK1,2/ERK1,2 pathway by either serum starvation or treatment with selective MEK1,2 inhibitors PD98059 and U0126 or expression of a dominant-negative mutant form of MEK1 decreased the amplitude of the G17-stimulated Ca2+ release response. Activation of the MEK1,2/ERK1,2 pathway either by pretreating cells with EGF or by expression of constitutively active K-ras (K-rasV12G) or MEK1 (MEK1*) increased the amplitude of G17-stimulated Ca2+ release. Although EGF, MEK1*, and K-rasV12G activated the MEK1,2/ERK1,2 pathway, they did not increase [Ca2+](i) in the absence of G17. These data demonstrate that the activation state of the MEK1,2/ERK1,2 pathway can modulate the amplitude of the CCK(2)R-mediated Ca2+ release response and identify a novel mechanism for cross-talk between EGF receptor- and CCK(2)R-regulated signaling pathways.


Assuntos
Cálcio/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Receptores da Colecistocinina/fisiologia , Animais , Ativação Enzimática , Gastrinas/farmacologia , Humanos , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Ratos
10.
Am J Physiol Lung Cell Mol Physiol ; 283(2): L364-72, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12114198

RESUMO

Airway epithelial cells represent the primary cell target of respiratory syncytial virus (RSV) infection. They actively participate in the lung immune/inflammatory response that follows RSV infection by expressing chemokines, small chemotactic cytokines that recruit and activate leukocytes. Regulated on activation, normal T cell expressed, and presumably secreted (RANTES) is a member of the CC chemokine subfamily and is strongly chemotactic for T lymphocytes, monocytes, basophils, and eosinophils, cell types that are present or activated in the inflammatory infiltrate that follows RSV infection of the lung. RSV infection of airway epithelial cells induces RANTES expression by increasing gene transcription and stabilizing RNA transcripts. The signaling pathway regulating RANTES gene expression after RSV infection has not been determined. In this study, we examined the role of extracellular signal-regulated kinase (ERK) and p38, members of the mitogen-activated protein (MAP) kinase (MAPK) family, in RSV-induced RANTES production. RSV infection of alveolar epithelial cells induced increased phosphorylation and catalytic activity of ERK and the upstream kinases Raf-1 and MAP ERK kinase. Induction of the MAP signaling cascade required a replication-competent virus. RSV infection of alveolar epithelial cells also induced activation of p38 MAPK. Inhibition of ERK and p38 activation significantly reduced RSV-induced RANTES mRNA and protein secretion without affecting RANTES gene transcription or transcription factor activation. These results indicate that the MAPK signaling cascade regulates RANTES production in alveolar epithelial cells through a posttranscriptional mechanism.


Assuntos
Quimiocina CCL5/genética , Regulação da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinase 1 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Processamento de Proteína Pós-Traducional , Infecções por Vírus Respiratório Sincicial/genética , Brônquios/citologia , Brônquios/metabolismo , Linhagem Celular , Quimiocina CCL5/metabolismo , Ativação Enzimática/fisiologia , Células Epiteliais/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/fisiologia , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA