Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
BMC Cancer ; 20(1): 707, 2020 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727409

RESUMO

BACKGROUND: A number of studies have reported hyperprogressive disease (HPD) in non-small cell lung cancer (NSCLC) after treatment with immune checkpoint inhibitor (ICI). This study aimed to summarize the incidence and survival outcome of HPD in NSCLC and identify the clinicopathological features associated with HPD based on available eligible studies. METHODS: Four databases (Medline/PubMed, Embase, Web of Science, and Cochrane Library) were searched for eligible studies on HPD published before January 23, 2020, to evaluate the incidence, outcome, and clinical features of HPD. Statistical analyses were performed using STATA 15.0. All meta-analyses were performed based on the random-effects model. RESULTS: This study included 6 studies involving 1389 patients. The incidence of HPD ranged from 8.02 to 30.43%. Compared with patients with non-HPD, those with HPD were associated with worse overall survival. We identified that Eastern Cooperative Oncology Group > 1, Royal Marsden Hospital score ≥ 2, serum lactate dehydrogenase > upper limit of normal, the number of metastasis sites > 2, and liver metastasis were associated with the risk of HPD. CONCLUSIONS: This study summarized the clinical features of HPD in NSCLC patients. The meta-analysis showed that five pre-treatment clinicopathological features might be associated with HPD, which may help in selecting patients for ICIs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Progressão da Doença , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia Adotiva/métodos , Neoplasias Pulmonares/terapia , Idoso , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/secundário , Feminino , Humanos , L-Lactato Desidrogenase/sangue , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
2.
Tumour Biol ; 37(9): 12223-12230, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27235120

RESUMO

IL-17 and IL-22 are linked to the development of intestinal inflammation and colorectal cancer (CRC). However, the maintenance of IL-17 and IL-22 production, as well as the cell type (Th17) that mediates these cytokines in CRC patients, remains unknown. To examine this, untreated CRC patients and healthy controls were recruited in this study. We first observed that CRC patients contained significantly elevated levels of IL-17- and IL-22-producing CD4+ T cells. The vast majority of IL-22-expressing CD4+ T cells also expressed IL-17. We then found that the production of both IL-17 and IL-22 required support from autologous monocytes, since the depletion of monocytes significantly downregulated IL-17 and IL-22 secretion. Naive T cells from CRC patients did not secrete IL-17 or IL-22 initially, but long-term coculture with autologous monocytes significantly upregulated IL-17 and IL-22 production in an IL-6-dependent manner. Blockade of IL-6 significantly reduced the levels of both IL-17 and IL-22. We then observed that CD163+ M2 macrophages were the main contributor of IL-6. Interestingly, incubation of monocytes with CCR4+CCR6+ Th17 cells resulted in significantly higher levels of CD163+ macrophages as well as higher IL-6 secretion, than incubation with non-Th17 CD4+ T cells. Together, our study discovered a positive feedback mechanism between Th17 and M2 macrophages in CRC patients.


Assuntos
Neoplasias Colorretais/metabolismo , Retroalimentação Fisiológica , Macrófagos/metabolismo , Células Th17/metabolismo , Idoso , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Técnicas de Cocultura , Neoplasias Colorretais/patologia , Feminino , Citometria de Fluxo , Humanos , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucinas/metabolismo , Macrófagos/classificação , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Receptores CCR4/metabolismo , Receptores CCR6/metabolismo , Receptores de Superfície Celular/metabolismo , Interleucina 22
3.
World J Microbiol Biotechnol ; 32(12): 192, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27718147

RESUMO

A post-fire morel collected from Populus simonii stands in Mt. Qingling was identified as Morchella crassipes Mes-20 by using nuclear ribosomal DNA internal transcribed spacer phylogeny. It was inoculated into sweet corn to observe colonized roots in purified culture and in greenhouse experiments. The elongation and maturation zones of sweet corn were remarkably colonized at the cortex intercellular and intracellular cells, vessel cells, and around the Casparian strip, forming ectendomycorrhiza-like structures. Colonization was also observed in the zone of cell division proximal to the root cap. Greenhouse assays with sweet corn showed that this morel stimulated the development of the root system and significantly increased the dry root biomass. M. crassipes also significantly reduced the incidence of Fusarium verticillioides in the kernels of mature ears when inoculated into young ears before Fusarium inoculation and prevented Fusarium infection in corn ears compared with that of the control in the greenhouse. When grown under axenic conditions, M. crassipes produced the phytohormones abscisic acid, indole-3-acetic acid, and salicylic acid. The benefits to plants elicited by M. crassipes may result from these phytohormones which may improve the drought resistance, biomass growth and resistance to Fusarium.


Assuntos
Doenças das Plantas/prevenção & controle , Reguladores de Crescimento de Plantas/farmacologia , Saccharomycetales/fisiologia , Zea mays/crescimento & desenvolvimento , Ácido Abscísico/metabolismo , Ácido Abscísico/farmacologia , Biomassa , Resistência à Doença , Fusarium/fisiologia , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacologia , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/microbiologia , Populus/microbiologia , Saccharomycetales/isolamento & purificação , Ácido Salicílico/metabolismo , Ácido Salicílico/farmacologia , Zea mays/microbiologia
4.
Gastroenterology ; 139(6): 2183-2194.e5, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20600029

RESUMO

BACKGROUND & AIMS: iASPP is an inhibitory member of the ankyrin-repeat-, SH3-domain- and proline-rich-region-containing protein (ASPP) family; iASPP expression is up-regulated in different human tumor types. We explored the molecular mechanism increased expression of iASPP and its role in hepatocellular carcinoma (HCC). METHODS: iASPP expression levels in human liver samples and cell lines were determined by polymerase chain reaction, immunoblot, and immunohistochemical analyses. Luciferase reporter, chromatin immunoprecipitation, and electrophoretic mobility shift assays were used to measure transcriptional activation by nuclear factor-κB (NF-κB). Effects on tumor growth were characterized with MTS, soft agar colony formation, and flow cytometry analyses. Tumorigenicity of cells was studied in nude mice. RESULTS: Compared with normal liver cells or tissues, iASPP was expressed at significantly higher levels in HCC cell lines (9/14) and liver samples from patients with HCC, cirrhosis, or hepatitis B virus infection. Increased expression of iASPP was significantly associated with time to recurrence and survival time of patients with HCC. NF-κB activation increased the expression of iASPP through p65/p50 binding to a putative NF-κB-binding site in the iASPP promoter; hepatitis B virus X gene product might up-regulate expression of iASPP. Transgenic expression of iASPP promoted tumor cell proliferation and resistance to chemotherapeutic drugs in vitro and in vivo. CONCLUSIONS: iASPP is up-regulated in HCC; it is a direct transcription target of NF-κB. Increased iASPP expression contributes to tumor progression by proliferative and antiapoptotic effects. iASPP might be developed as an HCC therapeutic target or to sensitize cancer cells to chemotherapeutic drugs; it might also be used as a prognostic factor.


Assuntos
Carcinoma Hepatocelular , Hepatite B Crônica/complicações , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas , NF-kappa B/metabolismo , Proteínas Repressoras/genética , Transativadores/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Prognóstico , Regiões Promotoras Genéticas/fisiologia , Transcrição Gênica/fisiologia , Proteínas Virais Reguladoras e Acessórias , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hepatology ; 51(1): 142-53, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20034025

RESUMO

UNLABELLED: The ankyrin-repeat-containing, SH3-domain-containing, and proline-rich-region-containing protein (ASPP) family of proteins regulates apoptosis through interaction with p53 and its family members. This study evaluated the epigenetic regulation of ASPP1 and ASPP2 in hepatitis B virus (HBV)-positive hepatocellular carcinoma (HCC) and explores the effects of down-regulation of ASPP1 and ASPP2 on the development of HCC. HCC cell lines and tissues from HCC patients were used to examine the expression and methylation of ASPP1 and ASPP2. The expression of ASPP1 and ASPP2 was diminished in HCC cells by epigenetic silence owing to hypermethylation of ASPP1 and ASPP2 promoters. Analyses of 51 paired HCC and surrounding nontumor tissues revealed that methylation of ASPP1 and ASPP2 was associated with the decreased expression of ASPP1 and ASPP2 in tumor tissues and the early development of HCC. Moreover, ASPP2 became methylated upon HBV x protein (HBx) expression. The suppressive effects on tumor growth by ASPP1 and ASPP2 were examined with RNA interference-mediated gene silence. Down-regulation of ASPP1 and ASPP2 promoted the growth of HCC cells in soft agar and in nude mice and decreased the sensitivity of HCC cells to apoptotic stimuli. CONCLUSION: ASPP1 and ASPP2 genes are frequently down-regulated by DNA methylation in HBV-positive HCC, which may play important roles in the development of HCC. These findings provide new insight into the molecular mechanisms leading to hepatocarcinogenesis and may have potent therapeutic applications.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo , Epigênese Genética , Vírus da Hepatite B/genética , Humanos , Neoplasias Hepáticas/virologia
6.
Int J Mol Med ; 42(1): 589-596, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29693157

RESUMO

Myocardial ischemia­reperfusion (I/R) injury is a major cause of cardiovascular disease worldwide, and microRNAs have been implicated in the regulation of pathological and physiological processes in myocardial I/R injury. The present study aimed to investigate the role of microRNA (miR)­221­3p in myocardial I/R injury. Cell death and lactate dehydrogenase (LDH) activity were increased in hydrogen peroxide (H2O2)­treated H9c2 cells, as measured by flow cytometry and an LDH detection kit. The expression of miR­221­3p was elevated in H2O2­incubated cells and in remote areas of the rat I/R model, examined using reverse transcription­quantitative polymerase chain reaction analysis. The overexpression of miR­221­3p enhanced the number of propidium iodide (PI)+ cells and the activity of LDH in H2O2­treated cells. In I/R­induced rats, the overexpression of miR­221­3p promoted the number of myosin+ cells and inhibited the fractional shortening of left ventricular diameter (FSLVD%). The results showed that the expression of p57 at the gene and protein levels was decreased in H9c2 cells incubated with H2O2 and in rats subjected to I/R surgery; the expression of p57 decreased following the overexpression of miR­221­3p. Subsequently, the hypothesis that p57 was the direct target of miR­221­3p was confirmed by performing a dual­luciferase reporter assay. Finally, to examine the function of p57 in myocardial impairment, p57 was transfected into H9c2 cells and administered to the rats prior to undergoing H2O2 treatment and I/R surgery, respectively. The results indicated that p57 attenuated the number of PI+ cells and the activity of LDH in H2O2­treated cells, whereas p57 downregulated the number of myosin+ cells and upregulated FSLVD% in the I/R­treated rats. Therefore, these findings suggested that miR­221­3p exacerbated the H2O2­induced myocardial damage in H9c2 cells and myocardial I/R injury in the rat model by modulating p57.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Peróxido de Hidrogênio/farmacologia , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Animais , Sequência de Bases , Linhagem Celular , Modelos Animais de Doenças , Masculino , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/cirurgia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos Sprague-Dawley , Regulação para Cima/genética
7.
Oncotarget ; 8(4): 5735-5752, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-27974696

RESUMO

Radiotherapy for esophageal squamous cell carcinoma (ESCC) patients is limited by resistance to ionizing radiation (IR). However, the roles and mechanisms of microRNAs in radioresistance are obscure. Here, we investigated that microRNA-205 (miR-205) was upregulated in radioresistant (RR) ESCC cells compared with the parental cells. Overexpression of miR-205 promoted colony survival post-IR, whereas depletion of miR-205 sensitized ESCC cells to IR in vitro and in vivo. Further, we demonstrated that miR-205 promoted radioresistance by enhancing DNA repair, inhibiting apoptosis and activating epithelial-mesenchymal transition (EMT). Mechanistically, miR-205, upregulated post-IR, was demonstrated to be activated by Sp1 in parallel with its host gene, miR-205HG, both of which showed a perfect correlation. We also identified and validated phosphatase and tensin homolog (PTEN), as a target of miR-205 that promoted radioresistance via PI3K/AKT pathway. Lastly, increased miR-205 expression was closely associated with decreased PTEN expression in ESCC tissues and miR-205 expression predicted poor prognosis in patients with ESCC. Taken together, these findings identify miR-205 as a critical determinant of radioresistance and a biomarker of prognosis. The Sp1-mediated transcriptional activation of miR-205 promotes radioresistance through PTEN via PI3K/AKT pathway in ESCC. Inhibition of miR-205 expression may be a new strategy for radiotherapy in ESCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Tolerância a Radiação , Fator de Transcrição Sp1/genética , Ativação Transcricional , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Transplante de Neoplasias , Prognóstico , Regiões Promotoras Genéticas , Transdução de Sinais , Análise de Sobrevida
8.
Oncotarget ; 6(4): 2120-33, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25544767

RESUMO

TGF-ß1, a potent EMT (epithelial-mesenchymal transition) inducer present in the tumor microenvironment, is involved in the metastasis and progression of various carcinomas, including esophageal squamous cell carcinoma (ESCC). TIP30 (30kDa HIV-1 Tat interacting protein) is a putative tumor metastasis suppressor. Here, we found TIP30 was decreased in cells undergoing EMT induced by TGF-ß1, an occurrence that was related to promoter hypermethylation. TGF-ß1 induced TIP30 hypermethylation via increasing DNMT1 and DNMT3A expression, which could be restored by TGF-ß antibodies. In our in vitro and in vivo studies, we showed that silence of TIP30 led to EMT, enhanced migrative and invasive abilities of ESCC cells, promoted tumor metastasis in xenografted mice; alternatively, overexpression of TIP30 inhibited TGF-ß1-induced EMT, and metastatic abilities of ESCC cells. Mechanically, TIP30 silencing induced the nuclear translocation and transcriptional activation of ß-catenin in an AKT-dependent manner, which further resulted in the initiation of EMT. Consistently, TIP30 was frequently methylated and downregulated in ESCC patients. Loss of TIP30 correlated with nuclear ß-catenin and aberrant E-cadherin expression. TIP30 was a powerful marker in predicting the prognosis of ESCC. Taken together, our results suggest a novel and critical role of TIP30 involved in TGF-ß1-induced activation of AKT/ß-catenin signaling and ESCC metastasis.


Assuntos
Acetiltransferases/genética , Carcinoma de Células Escamosas/genética , Metilação de DNA/efeitos dos fármacos , Neoplasias Esofágicas/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1/farmacologia , Acetiltransferases/metabolismo , Idoso , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Epigênese Genética/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Interferência de RNA , Análise de Sobrevida , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Transplante Heterólogo , beta Catenina/genética , beta Catenina/metabolismo
9.
Int J Clin Exp Pathol ; 7(7): 4120-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25120791

RESUMO

Regulator of G-protein signaling 6 (RGS6), a member of a family of RGS proteins, has been reported to involve in multiple processes during tumor development. However, its role in pancreatic cancer has not been studied yet. In this study, we aimed to investigate the expression of RGS6 in pancreatic cancer and its role in predicting outcomes of patients with pancreatic cancer. We first measured the expression of RGS6 mRNA in 20 cases of tumor tissues and matched adjacent non-tumorous tissues by quantitative real-time PCR and examined RGS6 protein by immunohistochemistry in tissue microarrays containing 90 tumor and 90 paired adjacent non-tumor tissues. Decreased RGS6 mRNA detected in primary tumor, compared with their non-tumor counterparts. In addition, decreased RGS6 protein expression was associated with tumor differentiation (P = 0.027), pT classification (P = 0.034), smoking status (P = 0.041) and a poor survival (P = 0.007). Cox proportional hazards regression modeling analysis revealed that lymph node metastasis (P = 0.001; hazard ratio, 2.347, 95% CI, 1.387-3.972), tumor differentiation (P = 0.015; hazard ratio, 0.505, 95% CI, 0.291-0.876) and RGS6 expression (P = 0.048; hazard ratio, 0.567, 95% CI, 0.324-0.994) were three independent prognostic factors. Taken together, these date demonstrate that RGS6 decreases in tumor tissue and may serve as a novel biomarker for outcomes in pancreatic cancer patients and be a potential therapeutic target potential therapeutic target.


Assuntos
Adenocarcinoma/patologia , Biomarcadores Tumorais/análise , Neoplasias Pancreáticas/patologia , Proteínas RGS/biossíntese , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Idoso , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Proteínas RGS/análise , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
10.
Nat Cell Biol ; 16(11): 1092-104, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25344754

RESUMO

Epithelial to mesenchymal transition (EMT), and the reverse mesenchymal to epithelial transition (MET), are known examples of epithelial plasticity that are important in kidney development and cancer metastasis. Here we identify ASPP2, a haploinsufficient tumour suppressor, p53 activator and PAR3 binding partner, as a molecular switch of MET and EMT. ASPP2 contributes to MET in mouse kidney in vivo. Mechanistically, ASPP2 induces MET through its PAR3-binding amino-terminus, independently of p53 binding. ASPP2 prevents ß-catenin from transactivating ZEB1, directly by forming an ASPP2-ß-catenin-E-cadherin ternary complex and indirectly by inhibiting ß-catenin's N-terminal phosphorylation to stabilize the ß-catenin-E-cadherin complex. ASPP2 limits the pro-invasive property of oncogenic RAS and inhibits tumour metastasis in vivo. Reduced ASPP2 expression results in EMT, and is associated with poor survival in hepatocellular carcinoma and breast cancer patients. Hence, ASPP2 is a key regulator of epithelial plasticity that connects cell polarity to the suppression of WNT signalling, EMT and tumour metastasis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Metástase Neoplásica , Fosforilação , Homeobox 1 de Ligação a E-box em Dedo de Zinco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa