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.
J Hematol Oncol ; 12(1): 112, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31703598

RESUMO

BACKGROUND: Increased liver stiffness exerts a detrimental role in driving hepatocellular carcinoma (HCC) malignancy and progression, and indicates a high risk of unfavorable outcomes. However, it remains largely unknown how liver matrix stiffness as an independent cue triggers epithelial-mesenchymal transition (EMT) and facilitates HCC metastasis. METHODS: Buffalo rat HCC models with different liver stiffness backgrounds and an in vitro Col I-coated cell culture system with tunable stiffness were used in the study to explore the effects of matrix stiffness on EMT occurrence and its underlying molecular mechanism. Clinical significance of liver stiffness and key molecules required for stiffness-induced EMT were validated in HCC cohorts with different liver stiffness. RESULTS: HCC xenografts grown in higher stiffness liver exhibited worse malignant phenotypes and higher lung metastasis rate, suggesting that higher liver stiffness promotes HCC invasion and metastasis. Cell tests in vitro showed that higher matrix stiffness was able to strikingly strengthen malignant phenotypes and independently induce EMT occurrence in HCC cells, and three signaling pathways converging on Snail expression participated in stiffness-mediated effect on EMT including integrin-mediated S100A11 membrane translocation, eIF4E phosphorylation, and TGF ß1 autocrine. Additionally, the key molecules required for stiffness-induced EMT were highly expressed in tumor tissues of HCC patients with higher liver stiffness and correlated with poor tumor differentiation and higher recurrence. CONCLUSIONS: Higher matrix stiffness as an initiator triggers epithelial-mesenchymal transition (EMT) in HCC cells independently, and three signaling pathways converging on Snail expression contribute to this pathological process. This work highlights a significant role of biomechanical signal in triggering EMT and facilitating HCC invasion and metastasis.


Assuntos
Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Hepáticas/patologia , Invasividade Neoplásica/patologia , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Experimentais , Distribuição Aleatória , Ratos , Ratos Endogâmicos BUF , Estudos Retrospectivos , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
2.
J Exp Clin Cancer Res ; 37(1): 99, 2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29728125

RESUMO

BACKGROUND: Higher matrix stiffness affects biological behavior of tumor cells, regulates tumor-associated gene/miRNA expression and stemness characteristic, and contributes to tumor invasion and metastasis. However, the linkage between higher matrix stiffness and pre-metastatic niche in hepatocellular carcinoma (HCC) is still largely unknown. METHODS: We comparatively analyzed the expressions of LOX family members in HCC cells grown on different stiffness substrates, and speculated that the secreted LOXL2 may mediate the linkage between higher matrix stiffness and pre-metastatic niche. Subsequently, we investigated the underlying molecular mechanism by which matrix stiffness induced LOXL2 expression in HCC cells, and explored the effects of LOXL2 on pre-metastatic niche formation, such as BMCs recruitment, fibronectin production, MMPs and CXCL12 expression, cell adhesion, etc. RESULTS: Higher matrix stiffness significantly upregulated LOXL2 expression in HCC cells, and activated JNK/c-JUN signaling pathway. Knockdown of integrin ß1 and α5 suppressed LOXL2 expression and reversed the activation of above signaling pathway. Additionally, JNK inhibitor attenuated the expressions of p-JNK, p-c-JUN, c-JUN and LOXL2, and shRNA-c-JUN also decreased LOXL2 expression. CM-LV-LOXL2-OE and rhLOXL2 upregulated MMP9 expression and fibronectin production obviously in lung fibroblasts. Moreover, activation of Akt pathway contributed to LOXL2-induced fibronectin upregulation. LOXL2 in CM as chemoattractant increased motility and invasion of BMCs, implicating a significant role of LOXL2 in BMCs recruitment. Except that, CM-LV-LOXL2-OE as chemoattractant also increased the number of migrated HCC cells, and improved chemokine CXCL12 expression in lung fibroblasts. The number of HCC cells adhered to surface of lung fibroblasts treated with CM-LV-LOXL2-OE was remarkably higher than that of the control cells. These results indicated that the secreted LOXL2 facilitated the motility of HCC cells and strengthened CTCs settlement on the remodeled matrix "soil". CONCLUSION: Integrin ß1/α5/JNK/c-JUN signaling pathway participates in higher matrix stiffness-induced LOXL2 upregulation in HCC cells. The secreted LOXL2 promotes fibronectin production, MMP9 and CXCL12 expression and BMDCs recruitment to assist pre-metastatic niche formation.


Assuntos
Aminoácido Oxirredutases/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Quimiocina CXCL12/metabolismo , Fibronectinas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Linhagem Celular Tumoral , Humanos , Transdução de Sinais , Regulação para Cima
3.
Cancer Med ; 7(3): 820-830, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29383859

RESUMO

Transarterial chemoembolization (TACE) is the standard treatment for unresectable hepatocellular carcinoma (HCC). Hypoxia-induced angiogenesis by TACE is linked to treatment failure; however, whether the chemotherapeutic damage of TACE to HCC could increase tumor angiogenesis has not been explored. The molecular effects of chemotherapy-damaged HCC cells on the neo-angiogenesis were investigated in vitro and in vivo. The expression of growth differentiation factor 15 (GDF15) was significantly upregulated in HCC cells exposed to chemotherapeutic agents. GDF15 from chemotherapy-damaged HCC cells promoted the in vitro proliferation, migration, and tube formation of endothelial cells. The pro-angiogenic effect of GDF15 was through the activation of Src and its downstream AKT, MAPK, and NF-κB signaling, which was blocked by thalidomide. The use of thalidomide significantly attenuated the in vivo chemotherapy-damaged HCC cells-promoted angiogenesis in nude mice. In conclusion, the chemotherapeutic damage in TACE to HCC could promote tumor angiogenesis via the increased release of GDF15. Thalidomide could reverse these pro-angiogenic effects.


Assuntos
Carcinoma Hepatocelular/complicações , Quimioembolização Terapêutica/efeitos adversos , Fator 15 de Diferenciação de Crescimento/efeitos adversos , Neoplasias Hepáticas/complicações , Neovascularização Patológica/etiologia , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Quimioembolização Terapêutica/métodos , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus
4.
Cancer Sci ; 108(9): 1778-1786, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28699238

RESUMO

Aggravated behaviors of hepatocellular carcinoma (HCC) will occur after inadequate thermal ablation. However, its underlying mechanisms are not fully understood. Here, we assessed whether the increased matrix stiffness after thermal ablation could promote the progression of residual HCC. Heat-treated residual HCC cells were cultured on tailorable 3D gel with different matrix stiffness, simulating the changed physical environment after thermal ablation, and then the mechanical alterations of matrix stiffness on cell phenotypes were explored. Increased stiffness was found to significantly promote the proliferation of the heat-treated residual HCC cells when the cells were cultured on stiffer versus soft supports, which was associated with stiffness-dependent regulation of ERK phosphorylation. Heat-exposed HCC cells cultured on stiffer supports showed enhanced motility. More importantly, vitamin K1 reduced stiffness-dependent residual HCC cell proliferation by inhibiting ERK phosphorylation and suppressed the in vivo tumor growth, which was further enhanced by combining with sorafenib. Increased matrix stiffness promotes the progression of heat-treated residual HCC cells, proposing a new mechanism of an altered biomechanical environment after thermal ablation accelerates HCC development. Vitamin K1 plus sorafenib can reverse this protumor effect.


Assuntos
Carcinoma Hepatocelular/patologia , Matriz Extracelular/patologia , Neoplasias Hepáticas Experimentais/patologia , Animais , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Terapia Combinada , Progressão da Doença , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hipertermia Induzida , Neoplasias Hepáticas Experimentais/terapia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasia Residual , Células-Tronco Neoplásicas/fisiologia , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Compostos de Fenilureia/farmacologia , Transdução de Sinais , Sorafenibe , Vitamina K 1/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Sci Rep ; 7(1): 2164, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28526827

RESUMO

Some evidences show that residual tumor after thermal ablation will progress rapidly. However, its mechanisms remain unclear. Here, we assessed whether activated HSCs could regulate stem cell-like property of residual tumor after incomplete thermal ablation to promote tumor progression. Human HCC cell lines were exposed to sublethal heat treatment to simulate the peripheral zone of thermal ablation. After residual HCC cells were cultured with conditional medium (CM) from activated HSCs, parameters of the stem cell-like phenotypes were analyzed. Nude mice bearing heat-exposed residual HCC cells and HSCs were subjected to metformin treatment to thwarter tumor progression. CM from activated primary HSCs or LX-2 cells significantly induced the stem cell-like phenotypes of residual HCC cells after heat treatment. These effects were significantly abrogated by neutralizing periostin (POSTN) in the CM. POSTN regulated the stemness of heat-exposed residual HCC cells via activation of integrin ß1/AKT/GSK-3ß/ß-catenin/TCF4/Nanog signaling pathway. Metformin significantly inhibited in vivo progression of heat-exposed residual HCC via suppressing POSTN secretion and decreasing cancer stem cell marker expression. Our data propose a new mechanism of activated HSCs promoting the stemness traits of residual HCC cells after incomplete thermal ablation and suggest metformin as a potential drug to reverse this process.


Assuntos
Carcinoma Hepatocelular/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Estreladas do Fígado/metabolismo , Temperatura Alta , Neoplasias Hepáticas/metabolismo , Fenótipo , Animais , Biomarcadores , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Masculino , Metformina/farmacologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , beta Catenina
6.
J Exp Clin Cancer Res ; 35(1): 114, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27431384

RESUMO

BACKGROUND: It is well established that some irradiated liver non-parenchymal cells secrete pro-inflammatory cytokines to facilitate the development of radiation-induced liver disease. However, little is known on whether the irradiated hepatoma cells-mediated non-irradiated hepatocyte injury occurs in HCC patients. Here, we elucidated the roles of the irradiated hepatoma cells in driving non-irradiated hepatocyte injury and its underlying mechanism. METHODS: SMMC7721 cells were cultured and divided into irradiated (4-Gy X-ray, R) and non-irradiated (NR) groups. At 24th hour after irradiation, conditioned medium (CM) from these cultures was mixed with normal culture medium in specific proportions, and termed as 7721-R-CM and 7721-NR-CM. Following incubation with these CM compound, the biological characteristics of L02 cells related to liver cell injury including viability, apoptosis and liver dysfunction indices were comparatively analyzed. Simultaneously, the levels of proliferation- and apoptosis-related cytokines in irradiated and non-irradiated SMMC7721 cells were also measured. FasL as a cytokine with significantly differential expression, was selected to clarify its effects on L02 apoptosis. Subsequently, FasL expression following irradiation was examined in SMMC7721 and other HCC cells with varying malignant potentials, as well as in HCC tissues, the related mechanism of higher expression of FasL in irradiated HCC cells was further investigated. RESULTS: Apoptosis and liver dysfunction indices were all significantly enhanced in L02 cells treated with 7721-R-CM, whereas viability was suppressed, compared to those with 7721-NR-CM stimulation. FasL was identified as a leading differential cytokine in the irradiated SMMC7721 cells. Higher proportion of apoptosis was also found in L02 cells following FasL incubation. A recombinant Fas-Fc protein, which blocks Fas-FasL interaction, ameliorated 7721-R-CM-induced apoptosis in L02 cells. FasL was highly expressed in a dose-dependent manner, and peaked at the 24th hour post-irradiation in different HCC cells and their culture supernatant. Meanwhile, phosphorylation levels of JNK, ERK, Akt, and p38 were all upregulated significantly in irradiated HCC cells. But, only JNK inhibition was validated to block radiation-induced FasL expression in HCC cells. c-Jun, the target transcription factor of JNK, was also activated. CONCLUSION: In HCC cells, the JNK-c-Jun pathway plays an important role in mediating irradiation- induced FasL expression, which may be critical in determining non-irradiated hepatocyte injury.


Assuntos
Carcinoma Hepatocelular/radioterapia , Proteína Ligante Fas/metabolismo , Hepatopatias/etiologia , Neoplasias Hepáticas/radioterapia , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/imunologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Hepatopatias/imunologia , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/imunologia , Masculino , Fosforilação , Proteínas Proto-Oncogênicas c-jun/metabolismo
7.
Oncotarget ; 7(22): 32221-31, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27050147

RESUMO

Matrix stiffness as an important physical attribute of extracellular matrix exerts significant impacts on biological behaviors of cancer cells such as growth, proliferation, motility, metabolism and invasion. However, its influence on cancer stemness still remains elusive. Here, we explore whether matrix stiffness-mediated effects on stemness characteristics occur in HCC cells. As the substrate stiffness increased, HCC cells exhibited high proportion of cells with CD133(+)/EpCAM(+), high expression levels of CD133, EpCAM, Nanog and SOX2, greater self-renewing ability and oxaliplatin resistance. Simultaneously, their phosphorylation levels of Akt and mTOR, as well as p-4E-BP and SOX2 expressions were also obviously upregulated. Conversely, knockdown of integrin ß1 partially attenuated higher stiffness-mediated stemness characteristics in HCC cells, and reversed the phosphorylation levels of Akt and mTOR, and expressions of p-4E-BP and SOX2, suggesting that integrin ß1 may deliver higher stiffness signal into HCC cells and activate mTOR signaling pathway. Additionally, mTOR inhibitor suppressed the mTOR phosphorylation level and expression levels of p-4E-BP and SOX2 in HCC cells grown on higher stiffness substrate, as well as depressed their stemness properties significantly, favoring a regulating role of mTOR signaling pathway in matrix stiffness-mediated effects on stemness. In summary, matrix stiffness may be involved in the process of stemness regulation via activating integrin ß1/Akt/mTOR/SOX2 signaling pathway. To the best of our knowledge, this study first reveals a novel regulating pathway to direct the stemness characteristics in HCC cells.


Assuntos
Carcinoma Hepatocelular/metabolismo , Matriz Extracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Antineoplásicos/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Autorrenovação Celular , Resistencia a Medicamentos Antineoplásicos , Elasticidade , Matriz Extracelular/patologia , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Mecanotransdução Celular , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Fatores de Transcrição SOXB1/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Transfecção
8.
PLoS One ; 10(8): e0134243, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26280346

RESUMO

Increased stromal stiffness is associated with hepatocellular carcinoma (HCC) development and progression. However, the molecular mechanism by which matrix stiffness stimuli modulate HCC progress is largely unknown. In this study, we explored whether matrix stiffness-mediated effects on osteopontin (OPN) expression occur in HCC cells. We used a previously reported in vitro culture system with tunable matrix stiffness and found that OPN expression was remarkably upregulated in HCC cells with increasing matrix stiffness. Furthermore, the phosphorylation level of GSK3ß and the expression of nuclear ß-catenin were also elevated, indicating that GSK3ß/ß-catenin pathway might be involved in OPN regulation. Knock-down analysis of integrin ß1 showed that OPN expression and p-GSK3ß level were downregulated in HCC cells grown on high stiffness substrate compared with controls. Simultaneously, inhibition of GSK-3ß led to accumulation of ß-catenin in the cytoplasm and its enhanced nuclear translocation, further triggered the rescue of OPN expression, suggesting that the integrin ß1/GSK-3ß/ß-catenin pathway is specifically activated for matrix stiffness-mediated OPN upregulation in HCC cells. Tissue microarray analysis confirmed that OPN expression was positively correlated with the expression of LOX and COL1. Taken together, high matrix stiffness upregulated OPN expression in HCC cells via the integrin ß1/GSK-3ß/ß-catenin signaling pathway. It highlights a new insight into a pathway involving physical mechanical signal and biochemical signal molecules which contributes to OPN expression in HCC cells.


Assuntos
Carcinoma Hepatocelular/metabolismo , Matriz Extracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Osteopontina/metabolismo , Transdução de Sinais , Regulação para Cima , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Integrina beta1/metabolismo , Fosforilação , Análise Serial de Tecidos , beta Catenina/metabolismo
9.
PLoS One ; 9(3): e88543, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24614035

RESUMO

BACKGROUND: Tumor cell invasion into the surrounding matrix has been well documented as an early event of metastasis occurrence. However, the dynamic expression patterns of proteins during early invasion of hepatocellular carcinoma (HCC) are largely unknown. Using a three-dimensional HCC invasion culture model established previously, we investigated the dynamic expression patterns of identified proteins during early invasion of HCC. MATERIALS AND METHODS: Highly metastatic MHCC97H cells and a liver tissue fragment were long-term co-cultured in a rotating wall vessel (RWV) bioreactor to simulate different pathological states of HCC invasion. The established spherical co-cultures were collected on days 0, 5, 10, and 15 for dynamic expression pattern analysis. Significantly different proteins among spheroids at different time points were screened and identified using quantitative proteomics of iTRAQ labeling coupled with LC-MS/MS. Dynamic expression patterns of differential proteins were further categorized by K-means clustering. The expression modes of several differentially expressed proteins were confirmed by Western blot and qRT-PCR. RESULTS: Time course analysis of invasion/metastasis gene expressions (MMP2, MMP7, MMP9, CD44, SPP1, CXCR4, CXCL12, and CDH1) showed remarkable, dynamic alterations during the invasion process of HCC. A total of 1,028 proteins were identified in spherical co-cultures collected at different time points by quantitative proteomics. Among these proteins, 529 common differential proteins related to HCC invasion were clustered into 25 types of expression patterns. Some proteins displayed significant dynamic alterations during the early invasion process of HCC, such as upregulation at the early invasion stage and downregulation at the late invasion stage (e.g., MAPRE1, PHB2, cathepsin D, etc.) or continuous upregulation during the entire invasion process (e.g., vitronectin, Met, clusterin, ICAM1, GSN, etc.). CONCLUSIONS: Dynamic expression patterns of candidate proteins during the early invasion process of HCC facilitate the discovery of new molecular targets for early intervention to prevent HCC invasion and metastasis.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Neoplasias/metabolismo , Proteômica/métodos , Western Blotting , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Análise por Conglomerados , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos , Humanos , Marcação por Isótopo , Neoplasias Hepáticas/genética , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proibitinas , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem
10.
Biochem Biophys Res Commun ; 444(3): 427-32, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24472554

RESUMO

Matrix stiffness as a novel regulation factor involves in modulating the pathogenesis of hepatocellular carcinoma (HCC) invasion or metastasis. However, the mechanism by which matrix stiffness modulates HCC angiogenesis remains unknown. Here, using buffalo rat HCC models with different liver matrix stiffness backgrounds and an in vitro cell culture system of mechanically tunable Collagen1 (COL1)-coated polyacrylamide gel, we investigated the effects of different matrix stiffness levels on vascular endothelial growth factor (VEGF) expression in HCC cells and explored its regulatory mechanism for controlling HCC angiogenesis. Tissue microarray analysis showed that the expression levels of VEGF and CD31 were gradually upregulated in tumor tissues with increasing COL1 and lysyl oxidase (LOX) expression, indicating a positive correlation between tumor angiogenesis and matrix rigidity. The expression of VEGF and the phosphorylation levels of PI3K and Akt were all upregulated in HCC cells on high-stiffness gel than on low-stiffness gel. Meanwhile, alteration of intergrin ß1 expression was found to be the most distinctive, implying that it might mediate the response of HCC cells to matrix stiffness simulation. After integrin ß1 was blocked in HCC cells using specific monoclonal antibody, the expression of VEGF and the phosphorylation levels of PI3K and Akt at different culture times were accordingly suppressed and downregulated in the treatment group as compared with those in the control group. All data suggested that the extracellular matrix stiffness stimulation signal was transduced into HCC cells via integrin ß1, and this signal activated the PI3K/Akt pathway and upregulated VEGF expression. This study unveils a new paradigm in which matrix stiffness as initiators to modulate HCC angiogenesis.


Assuntos
Matriz Extracelular/metabolismo , Integrina beta1/fisiologia , Neoplasias Hepáticas Experimentais/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Colágeno Tipo I/metabolismo , Ativação Enzimática , Lipoxigenase/metabolismo , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Neoplasias Hepáticas Experimentais/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos BUF
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...