Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Appl Immunohistochem Mol Morphol ; 29(7): 519-526, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33605573

RESUMO

BACKGROUND: Ovarian cancer (OC) is the most lethal malignancy of all female cancers and lacks an effective prognostic biomarker. Serous ovarian cancer (SOC) is the most common OC histologic type. The expression and function of bile acid receptor, G-protein-coupled bile acid receptor-1 (GPBAR1), in tumor progression remains controversial, and its clinical significance in SOC is unclear. MATERIALS AND METHODS: In our study, we detected the expression of GPBAR1 in SOCs and normal ovarian tissues with quantitative real-time polymerase chain reaction and immunohistochemistry to detect its expression pattern. Moreover, the prognostic significance of GPBAR1 was investigated with univariate and multivariate analyses. The function of GPBAR1 in regulating SOC proliferation was studied and the underlying mechanism was investigated with experiments in vitro. RESULTS: GPBAR1 was overexpressed in SOCs compared with the normal ovarian tissues. In the 166 SOCs, subsets with low and high GPBAR1 accounted for 57.23% and 42.77%, respectively. Moreover, our results suggested that GPBAR1 expression was significantly associated with poor prognosis and can be considered as an independent prognostic biomarker. With experiments in vitro, we suggested that GPBAR1 promoted SOC proliferation by increasing Smad4 ubiquitination, which required the involvement of GPBAR1-induced ERK phosphorylation. CONCLUSIONS: GPBAR1 was overexpressed in SOC and predicted the poor prognosis of SOC. We showed that GPBAR1 promoted SOC proliferation by activating ERK and ubiquitining Smad4. Our results suggested that GPBAR1 was a supplement to better classify SOC on the basis of the molecular profile and that GPBAR1 may be a potential drug target of SOC.


Assuntos
Carcinoma Epitelial do Ovário , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas , Receptores Acoplados a Proteínas G/biossíntese , Proteína Smad4/biossíntese , Ubiquitinação , Idoso , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Estudos Retrospectivos
2.
Pancreas ; 49(9): 1153-1160, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32897998

RESUMO

OBJECTIVE: For many patients whose pancreatic ductal adenocarcinoma (PDAC) is locally advanced, neoadjuvant therapy has been proposed as a way to decrease tumor burden. Pancreatic ductal adenocarcinoma is generally thought to be resistant to chemotherapy and radiation, however, response to neoadjuvant therapy in PDAC has been described in a subset of patients. The SMAD4 status is considered to be an important molecular feature which distinguishes two subsets of PDAC, SMAD4-positive and -negative tumors. The objective of this study was to evaluate the neoadjuvant treatment response rate as well as compare the different clinicopathologic variables between SMAD4-positive and -negative tumors. METHODS: We analyzed the data of patients who underwent surgical resection for PDAC from 2009-2019. Our cohort from a single institution included 233 patients. RESULTS: Of the 233 cases, 143 (61.4%) were SMAD4-negative and 90 (38.6%) were SMAD4-positive. Overall, SMAD4-positive tumors with neoadjuvant therapy had better treatment response and better tumor regression scores. In addition, SMAD4-positive tumors had a significantly lower lymph node metastasis rate in both the neoadjuvant and nonneoadjuvant setting. CONCLUSIONS: Further characterization of the role of SMAD4 within the context of neoadjuvant therapy will lead to improved personalized therapeutic strategies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Proteína Smad4/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Estudos de Coortes , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Avaliação de Resultados em Cuidados de Saúde/métodos , Avaliação de Resultados em Cuidados de Saúde/estatística & dados numéricos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico
3.
Eur J Cell Biol ; 99(4): 151076, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32439219

RESUMO

Deregulation of TGF-ß signaling is intricately engrossed in the pathophysiology of pancreatic adenocarcinomas (PDACs). The role of TGF-ß all through pancreatic cancer initiation and progression is multifarious and somewhat paradoxical. TGF-ß plays a tumor suppressive role in early-stage pancreatic cancer by promoting apoptosis and inhibiting epithelial cell cycle progression, but incites tumor promotion in late-stage by modulating genomic instability, neo-angiogenesis, immune evasion, cell motility, and metastasis. Here, we provide evidences that Par-4 acts as one of the vital mediators to regulate TGF-ß/Smad4 pathway, wherein, Par-4 induction/over-expression induced EMT which was later culminated in to apoptosis in presence of TGF-ß via positive regulation of Smad4. Intriguingly, Par-4-/- cells were devoid of significant Smad4 induction compared to Par-4+/+ cells in presence of TGF-ß and ectopic Par-4 steadily augmented Smad4 expression by restoring TGF-ß/Smad4 axis in Panc-1 cells. Further, our FACS and western blotting results unveiled that Par-4 dragged the PDAC cells to G1 arrest in presence of TGF-ß byelevating p21 and p27 levels while attenuating Cyclin E and A levels and augmenting caspase 3 cleavage triggering lethal EMT. Through restoration of Smad4, we further establish that in BxPC3 cell line (Smad4-/-), Smad4 is essential for Par-4 to indulge TGF-ß dependent lethal EMT program. The mechanistic relevance of Par-4 mediated Smad4 activation was additionally validated by co-immunoprecipitation wherein disruption of NM23H1-STRAP interaction by Par-4 rescues TGF-ß/Smad4 pathway in PDAC and mediates the tumor suppressive role of TGF-ß, therefore serving as a vital cog to restore the apoptotic functions of TGF-ß pathway.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Trombina/metabolismo , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias Pancreáticas/patologia , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Receptores de Trombina/genética , Transdução de Sinais , Proteína Smad4/biossíntese , Proteína Smad4/genética , Regulação para Cima
4.
Cell Cycle ; 18(21): 2860-2875, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31599708

RESUMO

Malignant melanoma has a profound influence on populations around the world, with the underlying mechanisms controlling this disease yet to be fully identified. Hence, the current study aimed to investigate effects associated with VEPH1 on epithelial-mesenchymal transition (EMT), proliferation, invasion, migration and the apoptosis of human cutaneous melanoma (CM) cells through the TGF-ß signaling pathway. Microarray-based gene analysis was initially performed to screen the CM-related differentially expressed genes. The expression of VEPH1, TGF-ß signaling pathway- and EMT-related genes in CM tissues and cell lines was subsequently evaluated. Gain-of- and loss-of-function experiments were conducted to examine the effects of VEPH1 and the TGF-ß signaling pathway on the expression of EMT-related genes, cell proliferation, migration, invasion, cell cycle and apoptosis in vitro. Finally, tumor formation in nude mice was conducted. VEPH1 was lowly expressed and regulated the progression of CM with involvement in the TGF-ß signaling pathway. Human CM tissues were noted to activate the TGF-ß signaling pathway and EMT. A375 cells treated with overexpressed VEPH1 plasmids or/and TGF-ß signaling pathway inhibitor SB-431542 displayed diminished TGF-ß, SMAD4, Vimentin and N-cadherin expression while the expression of E-cadherin was elevated, accompanied by decreased cell proliferation, migration, invasion, inhibited cell cycle entry. However, si-VEPH1 or TGF-ß signaling pathway activator contributed to reverse results. Taken together, the key findings of the current study present evidence suggesting that VEPH1 protects against human CM by inhibiting the activation of the TGF-ß signaling pathway, highlighting its potential as a target for the prognosis and diagnosis of CM.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melanoma/patologia , Neoplasias Cutâneas/patologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Adulto , Idoso , Animais , Apoptose/fisiologia , Benzamidas/farmacologia , Caderinas/biossíntese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Dioxóis/farmacologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Prognóstico , Transdução de Sinais/fisiologia , Proteína Smad4/biossíntese , Vimentina/biossíntese
5.
Histopathology ; 75(4): 546-551, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31054158

RESUMO

AIMS: SMAD4 (DPC4) is a tumour suppressor gene that is dysregulated in various tumour types, particularly pancreaticobiliary and gastrointestinal carcinomas. Corresponding loss of protein expression has been reported in approximately 50% of pancreatic and 25% of colonic adenocarcinomas. In the evaluation of carcinoma of unknown primary site, immunohistochemical loss of SMAD4 expression is often used to suggest pancreaticobiliary origin, but there are limited data on the spectrum of SMAD4 expression in carcinomas of other sites. This study evaluates the frequency of SMAD4 loss in a large cohort of carcinomas from diverse anatomical sites. METHODS AND RESULTS: Immunohistochemistry for SMAD4 was performed on tissue microarrays or whole tissue sections of 1210 carcinomas from various organs: gastrointestinal tract, liver, pancreas/biliary tract, lung, breast, thyroid, kidney, ovary and uterus. Expression was considered lost when there was complete absence of staining in tumour cell nuclei, in the presence of intact staining in non-neoplastic cells. SMAD4 loss was seen in 58% of pancreatic adenocarcinomas, 27% of appendiceal adenocarcinomas, 19% of colorectal adenocarcinomas, 16% of cholangiocarcinomas, 10% of lung adenocarcinomas and <5% of oesophageal, breast, gastric and mucinous ovarian adenocarcinomas. All papillary thyroid, hepatocellular, non-mucinous ovarian, endometrial and renal cell carcinomas showed intact SMAD4 nuclear expression. CONCLUSION: In addition to pancreaticobiliary, appendiceal and colonic tumours, SMAD4 loss is also seen in a small subset of other carcinomas, specifically breast, lung, oesophageal and gastric adenocarcinomas, all of which are typically CK7-positive, similar to pancreaticobiliary carcinoma. Awareness of SMAD4 loss in these other carcinoma types is helpful in the evaluation of carcinomas of unknown or uncertain primary site.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma/metabolismo , Carcinoma/patologia , Proteína Smad4/biossíntese , Feminino , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Humanos , Masculino , Proteína Smad4/análise
6.
Mod Pathol ; 32(6): 844-854, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30683911

RESUMO

Metastasis following surgical resection is a leading cause of mortality in pancreatic ductal adenocarcinoma. Epithelial-mesenchymal transition is thought to play an important role in metastasis, although its clinical relevance in metastasis remains uncertain. We evaluated a panel of RNA in-situ hybridization probes for epithelial-mesenchymal transition-related genes expressed in circulating tumor cells. We assessed the predictive value of this panel for metastasis in pancreatic ductal adenocarcinoma and, to determine if the phenotype is generalizable between cancers, in colonic adenocarcinoma. One hundred fifty-eight pancreatic ductal adenocarcinomas and 205 colonic adenocarcinomas were classified as epithelial or quasimesenchymal phenotype using dual colorimetric RNA-in-situ hybridization. SMAD4 expression on pancreatic ductal adenocarcinomas was assessed by immunohistochemistry. Pancreatic ductal adenocarcinomas with quasimesenchymal phenotype had a significantly shorter disease-specific survival (P = 0.031) and metastasis-free survival (P = 0.0001) than those with an epithelial phenotype. Pancreatic ductal adenocarcinomas with SMAD4 loss also had lower disease-specific survival (P = 0.041) and metastasis-free survival (P = 0.001) than those with intact SMAD4. However, the quasimesenchymal phenotype proved a more robust predictor of metastases-area under the curve for quasimesenchymal = 0.8; SMAD4 = 0.6. The quasimesenchymal phenotype also predicted metastasis-free survival (P = 0.004) in colonic adenocarcinoma. Epithelial-mesenchymal transition defined two phenotypes with distinct metastatic capabilities-epithelial phenotype tumors with predominantly organ-confined disease and quasimesenchymal phenotype with high risk of metastatic disease in two epithelial malignancies. Collectively, this work validates the relevance of epithelial-mesenchymal transition in human gastrointestinal tumors.


Assuntos
Carcinoma Ductal Pancreático/patologia , Transição Epitelial-Mesenquimal , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/patologia , Adenocarcinoma/patologia , Idoso , Biomarcadores Tumorais/análise , Neoplasias Colorretais/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Proteína Smad4/biossíntese , Neoplasias Pancreáticas
7.
Artigo em Inglês | MEDLINE | ID: mdl-30595209

RESUMO

Color Index (C.I.) Disperse Blue 291 (DB291) is an azo dye used by the textile industry. After yarn dyeing, wastewater containing the dye, released into the aquatic environment, may pollute drinking water sources. We investigated the mutagenicity and genotoxicity of DB291 in male Swiss mice, following oral administration. Micronucleated cells, primary DNA damage (comet assay) in blood, liver, and kidney cells, and BAX, BCL2, SMAD4 and TNFA gene expression in leukocytes were evaluated. An increased frequency of micronucleated polychromatic erythrocytes (MNPCEs) was observed in animals treated with 50 mg/kg bw; no other genetic alteration was detected. Neither primary DNA damage nor changes in gene expression were observed.


Assuntos
Compostos Azo/toxicidade , Medula Óssea/efeitos dos fármacos , Corantes/toxicidade , Eritrócitos Anormais/efeitos dos fármacos , Animais , Compostos Azo/farmacologia , Ensaio Cometa , Dano ao DNA/genética , Expressão Gênica/efeitos dos fármacos , Leucócitos/metabolismo , Masculino , Camundongos , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteína Smad4/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Águas Residuárias/química
8.
Rom J Morphol Embryol ; 60(3): 803-809, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31912090

RESUMO

Pancreatic ductal carcinoma is the most common type of pancreatic cancer, and currently represents the fourth cause of death by cancer, worldwide. Among classical pancreatic markers that ascertain the histopathology, new emerging targets have been proposed for both diagnostic and prognostic purposes. In the present study, utilizing a group of 28 confirmed resected pancreatic ductal carcinomas, we have assessed the immunoexpression and correlation ratios of mothers against decapentaplegic homolog 4 (Drosophila) (SMAD4)∕transforming growth factor beta receptor 2 (TGFßR2), and vimentin∕cluster of differentiation 105 (CD105). SMAD4 showed an overall increase in tumors versus pancreatic control tissue, but a decrease from G1 towards poorly differentiated tumors, while TGFßR2, vimentin and CD105 showed higher expression values in the tumor areas. Vimentin-CD105 colocalization degree decreased in tumor tissues compared to controls, illustrating a desynchronization of these two markers, both of them being negative in the tumor epithelia. Altogether, it is highly plausible that all these key players revolve around the epithelial-to-mesenchymal transition phenomenon, and this itself modulates the clinical outcome of the patient.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/biossíntese , Proteína Smad4/biossíntese , Adulto , Idoso , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Transição Epitelial-Mesenquimal , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Proteína Smad4/genética
9.
Exp Hematol ; 68: 66-79.e3, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30208330

RESUMO

The selection of chemotherapy regimen for elderly patients with acute myeloid leukemia (AML) remains challenging. Here, we report that granulocyte colony-stimulating factor (G-CSF) upregulates the expression of microRNA (miR)-146a in a nuclear factor kappaB-dependent manner, leading to direct decreases in the expression of the target proteins CXCR4 and Smad4 in AML cells in vitro. The reduction in CXCR4 expression suppressed the migration abilities of leukemia cells. Downregulation of Smad4 promoted cell cycle entry in leukemia cells. Furthermore, an increase in apoptosis was observed when leukemia cells were treated sequentially with G-CSF and cytosine arabinoside in vitro. These findings suggest that G-CSF treatment may disrupt the protection of bone marrow niches from leukemia cells. In a review of data from 78 cases of primary AML, we found that a high miR-146a expression and/or upregulation of this miRNA during G-CSF priming chemotherapy was predictive of better clinical outcomes. Our findings suggest that miR-146a may be a novel biomarker for evaluating the clinical prognosis and treatment effects of a G-CSF priming protocol in elderly patients with AML.


Assuntos
Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , MicroRNAs/fisiologia , RNA Neoplásico/fisiologia , Aclarubicina/administração & dosagem , Aclarubicina/efeitos adversos , Aclarubicina/farmacologia , Fatores Etários , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiotaxia/efeitos dos fármacos , Técnicas de Cocultura , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Células HL-60 , Humanos , Leucemia Mielomonocítica Aguda/tratamento farmacológico , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , Interferência de RNA , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , RNA Interferente Pequeno/genética , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Proteína Smad4/biossíntese , Proteína Smad4/genética , Nicho de Células-Tronco , Microambiente Tumoral , Regulação para Cima/efeitos dos fármacos
10.
Mol Med Rep ; 17(5): 6397-6404, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29512734

RESUMO

The incidence of vulvar squamous cell carcinoma (VSCC) has increased annually over the last decade. MicroRNAs (miRNAs/miRs) serve an important role in tumor progression and development. Our previous microarray studies have revealed that miR­3147 was overexpressed in VSCC. However, its function and underlying mechanism in VSCC remain unknown. In the present study, it was confirmed by reverse transcription­quantitative polymerase chain reaction that the expression of miR­3147 was markedly upregulated in VSCC tissues. The increased expression of miR­3147 was positively associated with the depth of invasion. The overexpression of miR­3147 resulted in the promotion of vulvar cancer cell proliferation, migration, invasion, G1/S progression and invasion­associated gene expression. miR­3147 may participate in the process of epithelial­mesenchymal transition and reduce the expressions of downstream target genes in the transforming growth factor­ß/Smad signaling pathway in A431 cells. The knockdown of Smad4 by small interfering RNA promoted malignant behaviours in A431 cells. In addition, miR­3147 regulated Smad4 by directly binding to its 3' untranslated region. In conclusion, the results indicated that miR­3147 may serve an oncogenic role in VSCC by targeting Smad4. miR­3147 may represent a novel potential therapeutic target marker for VSCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/metabolismo , Proteína Smad4/biossíntese , Neoplasias Vulvares/metabolismo , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Feminino , Humanos , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Proteína Smad4/genética , Neoplasias Vulvares/genética
11.
Pancreas ; 47(2): 208-212, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29329157

RESUMO

OBJECTIVES: The tumor suppressor gene SMAD4 (DPC4) is genetically inactivated in approximately half of pancreatic ductal adenocarcinomas (PDAs). We examined whether Smad4 tumor status was associated with outcomes after adjuvant chemoradiation (CRT) for resected PDAs. METHODS: Patients treated with adjuvant CRT were identified (N = 145). Smad4 status was determined by immunolabeling and graded as intact or lost. Kaplan-Meier method and multivariable competing risk analyses were performed. RESULTS: On multivariate competing risk analysis, Smad4 loss was associated with increased risk of local recurrence (LR) (hazard ratio, 2.37; 95% confidence interval, 1.10-5.11; P = 0.027), distant failure (DF) (hazard ratio, 1.71; 95% confidence interval, 1.03-2.83; P = 0.037), and synchronous LR and DF at first recurrence (14.9 % vs 5.3%, P = 0.07) compared with Smad4 intact cancers. Smad4 loss was not associated with median overall survival (22 vs 22 months; P = 0.63) or disease-free survival (lost [13.6 months] vs intact [13.5 months], P = 0.79). CONCLUSIONS: After PDA resection and adjuvant CRT, Smad4 loss correlated with higher risk of LR and DF, but not with survival. Smad4 loss may help predict which surgical patients are at higher risk for failure after definitive management and may benefit from intensified adjuvant therapy.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma Ductal Pancreático/terapia , Neoplasias Pancreáticas/terapia , Proteína Smad4/biossíntese , Idoso , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/cirurgia , Quimiorradioterapia Adjuvante , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Mutação , Recidiva Local de Neoplasia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Estudos Retrospectivos , Proteína Smad4/genética , Resultado do Tratamento
12.
J Cachexia Sarcopenia Muscle ; 9(1): 119-128, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28984049

RESUMO

BACKGROUND: Loss of muscle mass and strength are important sequelae of chronic disease, but the response of individuals is remarkably variable, suggesting important genetic and epigenetic modulators of muscle homeostasis. Such factors are likely to modify the activity of pathways that regulate wasting, but to date, few such factors have been identified. METHODS: The effect of miR-422a on SMAD4 expression and transforming growth factor (TGF)-ß signalling were determined by western blotting and luciferase assay. miRNA expression was determined by qPCR in plasma and muscle biopsy samples from a cross-sectional study of patients with chronic obstructive pulmonary disease (COPD) and a longitudinal study of patients undergoing aortic surgery, who were subsequently admitted to the intensive care unit (ICU). RESULTS: miR-422a was identified, by a screen, as a microRNA that was present in the plasma of patients with COPD and negatively associated with muscle strength as well as being readily detectable in the muscle of patients. In vitro, miR-422a suppressed SMAD4 expression and inhibited TGF-beta and bone morphogenetic protein-dependent luciferase activity in muscle cells. In male patients with COPD and those undergoing aortic surgery and on the ICU, a model of ICU-associated muscle weakness, quadriceps expression of miR-422a was positively associated with muscle strength (maximal voluntary contraction r = 0.59, P < 0.001 and r = 0.51, P = 0.004, for COPD and aortic surgery, respectively). Furthermore, pre-surgery levels of miR-422a were inversely associated with the amount of muscle that would be lost in the first post-operative week (r = -0.57, P < 0.001). CONCLUSIONS: These data suggest that differences in miR-422a expression contribute to the susceptibility to muscle wasting associated with chronic and acute disease and that at least part of this activity may be mediated by reduced TGF-beta signalling in skeletal muscle.


Assuntos
MicroRNAs/metabolismo , Debilidade Muscular/metabolismo , Músculo Esquelético/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Proteína Smad4/metabolismo , Idoso , Linhagem Celular , Estudos de Coortes , Estudos Transversais , Feminino , Humanos , Masculino , MicroRNAs/biossíntese , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Debilidade Muscular/genética , Músculo Esquelético/metabolismo , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Pulmonar Obstrutiva Crônica/genética , Transdução de Sinais , Proteína Smad4/biossíntese , Transfecção , Fator de Crescimento Transformador beta/metabolismo
13.
Oncol Rep ; 38(4): 2317-2324, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28902368

RESUMO

Parthenolide (PT), a sesquiterpene lactone extracted from the plant feverfew, has been demonstrated to have anti-inflammatory and anticancer properties. Although PT has been revealed to markedly inhibit colorectal cancer cell proliferation, the inhibitory effects decrease with administration time. These findings revealed that colorectal cancer cells develop resistance to PT. However, the underlying mechanism is unclear. In the present study we observed significantly low expression of Smad4 in 3 PT-resistant cell lines (HCT­116/PT, HT-29/PT and Caco-2/PT), which were obtained using in vitro concentration gradient-increased induction, but not in their parental cells. In the present study we used the lentiviral­mediated transfection method to upregulate Smad4 in resistant colorectal cancer cell lines. Flow cytometry assay was used to assess cell apoptosis. Cell migration was detected using a QCM™ 24-well Fluorimetric Cell Migration Assay kit. Our study showed that Smad4 overexpression notably decreased the half maximal inhibitory concentration (IC50) values for PT in the 3 PT-resistant cell lines, and improved the inhibitory effects of PT on cell migration and enhanced apoptosis in vitro as well as suppressed xenografted tumors in a PT-resistant colorectal cancer mouse model. Further study by western blotting into the underlying mechanism demonstrated that Smad4 overexpression suppressed the expression of MDR1 in the resistant cells, and resulted in the accumulation of PT, which in turn promoted the expession of caspase-3 and Bax and inhibited the expression of Bcl-2 and the phosphorylation of NF-κB p65. In short, Smad4 re-expression may be crucial for enhancing the sensitivity and reversing the resistance to PT in PT-resistant colorectal cancer cells.


Assuntos
Neoplasias Colorretais/terapia , Sesquiterpenos/farmacologia , Proteína Smad4/biossíntese , Animais , Células CACO-2 , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos , Terapia Genética , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Distribuição Aleatória , Proteína Smad4/genética , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Tumour Biol ; 39(6): 1010428317705513, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28631567

RESUMO

Abnormal expression of microRNAs has been reported to regulate gene expression and cancer cell growth, invasion, and migration. Recently, upregulation of hsa-miR-1285 was demonstrated in bronchoalveolar lavage fluid samples from patients with lung cancer and downregulation in plasma level of stage-I lung cancer patients. However, the function and the underlying mechanism of miR-1285 in non-small-cell lung carcinoma have not been elucidated. In this study, we found that miR-1285-5p, the mature form of miR-1285, was significantly upregulated in human non-small-cell lung carcinoma cell lines A549 and SK-MES-1. Additionally, cells transfected with the miR-1285-5p inhibitor LV-anti-miR-1285-5p demonstrated significantly inhibited proliferation and invasion and depressed migration. Further analysis demonstrated that the miR-1285-5p precursor LV-miR-1285-5p attenuated the expression of Smad4 and cadherin-1 (CDH1) but that LV-anti-miR-1285-5p showed opposite results. A luciferase reporter assay confirmed that miR-1285-5p targeted Smad4 and CDH1. Mechanism analyses revealed that silence of Smad4 and CDH1 significantly attenuated the inhibitory effects of LV-anti-miR-1285-5p on non-small-cell lung carcinoma growth and invasion. Taken together, our data suggest that miR-1285-5p functions as a tumor promoter in the development of non-small-cell lung carcinoma by targeting Smad4 and CDH1, indicating a novel therapeutic strategy for non-small-cell lung carcinoma patients.


Assuntos
Caderinas/biossíntese , Carcinoma Pulmonar de Células não Pequenas/genética , MicroRNAs/genética , Proteína Smad4/biossíntese , Células A549 , Adulto , Idoso , Antígenos CD , Caderinas/genética , Carcinogênese/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteína Smad4/genética
15.
Int J Oncol ; 50(5): 1693-1700, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28393199

RESUMO

SMAD4 is a critical co-smad in signal transduction pathways activated in response to transforming growth factor-ß (TGF-ß)-related ligands, regulating cell growth and differentiation. The roles played by SMAD4 inactivation in tumors highlighted it as a tumor-suppressor gene. Herein, we report that loss of SMAD4 expression in vascular endothelial cells promotes ovarian cancer invasion. SiRNA transfer of this gene in the HUVEC reduced SMAD4 protein expression and function. Although it reduced the vessel endothelial cell tubule formation in vitro and in vivo, it did not affect the tumor growth significantly in vivo. However, it weakened the barrier integrity in endothelial cells and increased vessel permeability and the ovarian cancer liver metastasis. We documented reduced angiogenesis and increased invasion histologically and by intravital microscopy, and gained mechanistic insight at the messenger and gene level. Finally, we found a negative reciprocal regulation between SMAD4 and FYN. FYN is one of the Src family kinases (SFK), activation of which can cause dissociation of cell-cell junctions and adhesion, resulting in paracellular hypermeability. Upon SMAD4 deletion, we detected high expression levels of FYN in vessel endothelial cells, suggesting the mechanism of the ovarian tumor cells cross the endothelial barrier and transform to an invasive phenotype.


Assuntos
Neoplasias Hepáticas/genética , Neoplasias Ovarianas/genética , Proteínas Proto-Oncogênicas c-fyn/biossíntese , Proteína Smad4/biossíntese , Animais , Vasos Sanguíneos/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Células Endoteliais/patologia , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-fyn/genética , Proteína Smad4/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Tumour Biol ; 39(3): 1010428317694551, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28349818

RESUMO

The SMAD proteins are responsible for transducing signals from activated transforming growth factor-beta. This is the first study assessing the expression of SMAD-1/8, SMAD-2/3, SMAD-4, and SMAD-7 in chronic lymphocytic leukemia cells with regard to their clinical significance and potential prognostic value. Overexpression of SMAD-1/8 was observed in 160 chronic lymphocytic leukemia patients compared to 42 healthy volunteers (p = 0.023) and was associated with a more progressive course of the disease (p = 0.016). Moreover, the high expression of SMAD-1/8 correlated with other, well-established prognostic factors, including clinical stage (p = 0.010) and lymphocyte doubling time (p = 0.021). The expression of SMAD-4 was lower in chronic lymphocytic leukemia patients compared with the control group (p = 0.003). Importantly, lower SMAD-4 levels correlated with longer progression-free survival (p = 0.009), progressive course of the disease (p = 0.002), advanced clinical stage (p = 0.0004), elevated beta-2-microglobulin and lactate dehydrogenase levels (p < 0.05), shorter lymphocyte doubling time (p = 0.009), and CD38 antigen expression (p = 0.039). In addition, lower SMAD-4 expression correlated with lower apoptotic index (p = 0.0007) and lower expression of receptors for vascular endothelial growth factors VEGFR-1 and VEGFR-2. A significant association was found between the low expression of inhibitory protein SMAD-7 and both zeta-chain-associated protein kinase 70-negative cells (p = 0.04) and lower apoptotic index (p = 0.004). No differences were observed in SMAD-2/3 expression. In conclusion, our results demonstrate a significant correlation between greater SMAD-1/8 and lower SMAD-4 expression in chronic lymphocytic leukemia cells, as well as more progressive outcome and poor prognosis. These data provide supporting evidence that the expression of SMAD proteins plays an important role in disease development and may be considered as a novel, biologic prognostic factor in this disease.


Assuntos
Leucemia Linfocítica Crônica de Células B/genética , Proteína Smad1/biossíntese , Proteína Smad2/biossíntese , Proteína Smad4/biossíntese , Proteína Smad7/biossíntese , Idoso , Idoso de 80 Anos ou mais , Apoptose/genética , Intervalo Livre de Doença , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteína Smad1/genética , Proteína Smad2/genética , Proteína Smad4/genética , Proteína Smad7/genética , Fator de Crescimento Transformador beta/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Proteína-Tirosina Quinase ZAP-70/biossíntese
17.
Int J Oncol ; 50(1): 272-282, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27922669

RESUMO

SMAD4 is a tumor suppressor that is frequently inactivated in many types of cancer. The role of abnormal expression of SMAD4 has been reported in developmental processes and the progression of various human cancers. The expression level of SMAD4 has been related to the survival rate in gastric cancer patients. However, the molecular mechanism underlying transcriptional regulation of SMAD4 remains largely unknown. In the present study, we characterized the promoter region of SMAD4 and identified myeloid zinc finger 1 (MZF1), as a putative transcription factor. MZF1 directly bound to a core region of the SMAD4 promoter and stimulated transcriptional activity. We also found that the expression of MZF1 influences the migration ability of gastric adenocarcinoma cells. Collectively, our results showed that MZF1 has a role in cellular migration of gastric cancer cells via promoting an increase in intracellular SMAD4 levels. This study might provide new evidence for the molecular basis of the tumor suppressive effect of the MZF1-SMAD4 axis, a new therapeutic target in advanced human gastric cancer.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Regiões Promotoras Genéticas , Proteína Smad4/biossíntese , Neoplasias Gástricas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Neoplasias Gástricas/patologia , Ativação Transcricional/genética
18.
Int J Oncol ; 49(6): 2589-2599, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27779660

RESUMO

MicroRNAs (miRNAs) were reported to be involved in the development of clear cell renal cell carcinoma (ccRCC). However, the study on miRNAs in ccRCC is far from complete. The present study identified miRNAs which could act as potential novel prognostic markers for ccRCC, and analyzed its possible mechanism. We found that miR-19a correlated with poor prognosis of ccRCC patients via promoting cell proliferation and suppressing PTEN/SMAD4 expression. Both the microarray screening result and TCGA KIRC dataset analysis showed that miR-19a was significantly upregulated in ccRCC tissues, and further analysis of TCGA data revealed that the upregulated level of miR-19a was strongly associated with advanced T stage and poor prognosis of ccRCC patients. Consistent with clinical observations, miR-19a overexpression significantly promoted ccRCC cell proliferation in vitro. To further explore the mechanism by which miR-19a correlated with cell proliferation and poor prognosis of ccRCC, we performed gene set enrichment analysis (GSEA) for target genes of miR-19a in ccRCC patients. Result indicated that the key target genes of miR-19a included SMAD4 and PTEN. In ccRCC tissues, expression levels of SMAD4 and PTEN were negatively correlated with expression level of miR-19a, revealing that miR-19a suppressed the expression of SMAD4 and PTEN in ccRCC patients. miR-19a overexpression significantly suppressed the expression of SMAD4 and PTEN in vitro, further verifying that SMAD4 and PTEN were the target genes of miR-19a in ccRCC cells. Our results elucidated the tumor promoting role of miR-19a and established miR-19a as a potential novel prognostic marker for ccRCC.


Assuntos
Carcinoma de Células Renais/patologia , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Renais/patologia , MicroRNAs/genética , PTEN Fosfo-Hidrolase/biossíntese , Proteína Smad4/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/mortalidade , Linhagem Celular Tumoral , Regulação para Baixo/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/mortalidade , Prognóstico , Mapas de Interação de Proteínas
19.
Oncol Rep ; 36(4): 2357-64, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27498705

RESUMO

The contribution of aberrant osteopontin (OPN) expression to tumor progression and metastasis has been documented in a wide spectrum of malignancies, and targeted inhibition of OPN has therefore emerged as an attractive strategy for cancer therapy. Transcription of OPN is regulated by various transcription factors, and our recently published study demonstrated that downregulation of OPN is an important event in the TGF­ß cytostatic program. We report here that brefelamide exerts an inhibitory effect on OPN expression and function in A549 human lung carcinoma cells. The promoter, RNA, and protein levels of OPN were decreased in brefelamide­treated A549 cells, which was accompanied by reduced invasive ability in vitro. OPN inhibition by brefelamide was largely abrogated by disruption of a putative TGF­ß inhibitory element in the OPN promoter. Treatment with brefelamide induced Smad4 expression, and knockdown of Smad4 by RNA interference partially diminished the inhibitory effect of brefelamide on OPN. These results indicate that brefelamide inhibited OPN­mediated cell invasion through restoration of the OPN repression by TGF­ß/Smad signaling. Together with the reported antiproliferative property, our findings suggest that brefelamide might serve as a potential candidate for the development of a new antitumor and antimetastatic agent.


Assuntos
Amidas/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Invasividade Neoplásica/genética , Osteopontina/genética , Fenóis/administração & dosagem , Células A549 , Apoptose/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/patologia , Metástase Neoplásica , Osteopontina/antagonistas & inibidores , Osteopontina/biossíntese , Regiões Promotoras Genéticas , Interferência de RNA , Proteína Smad4/biossíntese , Proteína Smad4/genética
20.
Int J Oncol ; 49(2): 700-8, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27279345

RESUMO

The TGF-ß/Smad signaling pathway plays important roles in cancer cell proliferation, apoptosis, differentiation, angiogenesis and epithelial-mesenchymal transition (EMT), which is the key event in the early stages of cancer metastasis and enhances the capability of cell migration and invasion. Smad4 acts as the only Co-Smad of TGF/Smad signaling pathway and plays the key role in TGF-ß-mediated EMT. Nevertheless, the mRNA regulation mechanisms of Smad4 in human non-small cell lung cancer (NSCLC) remains largely unclear. Computational algorithms predicted that the 3'-UTR of Smad4 is a target of miR­205. Here, we validated that miR­205 could directly bind to 3'-UTR of Smad4 by luciferase assays. Moreover, we investigated the functional roles of miR­205 and its molecular link to Smad4 in lung cancer cells. In this study, we confirmed that overexpression of miR­205 suppressed the expression of Smad4, in turn, weakened the TGF-ß/Smad signaling pathway and inhibited TGF-ß/Smad4-induced EMT, invasion and migration ultimately. Furthermore, this study shows that miR­205 can serve as a promising therapeutic target of highly aggressive NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , MicroRNAs/genética , Proteína Smad4/genética , Fator de Crescimento Transformador beta/genética , Regiões 3' não Traduzidas , Células A549 , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/biossíntese , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Ligação Proteica , Transdução de Sinais/genética , Proteína Smad4/antagonistas & inibidores , Proteína Smad4/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA