Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 209(10): 1987-1998, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36426955

RESUMO

Pyroptosis is a form of regulated cell death mediated by the gasdermin protein family. During virus infection, cell pyroptosis restricts viral replication. The mechanisms of the tripartite motif (TRIM) protein family and IFN-stimulated genes (ISGs) against viruses have been studied. The role of TRIMs and ISGs in pyroptosis remains unclear. In this study, we show that TRIM21 interacts with ISG12a in viral infection and facilitates its translocation into the mitochondria by promoting its ubiquitination, thereby causing caspase 3 activation. Gasdermin E (GSDME) is specifically cleaved by caspase 3 upon viral infection, releasing the GSDME N-terminal domain, perforating the cell membrane, and causing cell pyroptosis. Our study uncovers a new mechanism of TRIM21 and ISG12a in regulating virus-induced cell pyroptosis.


Assuntos
Piroptose , Vírus , Piroptose/fisiologia , Caspase 3/metabolismo , Ubiquitinação , Morte Celular , Proteínas com Motivo Tripartido/metabolismo
2.
Int J Cancer ; 152(2): 110-122, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35765844

RESUMO

Pancreatic cancer (PC) is a cancer of the digestive system, and pancreatic ductal adenocarcinoma (PDAC) accounts for approximately 90% of all PC cases. Exosomes derived from PDAC (PDAC-exosomes) promote PDAC development and metastasis. Exosomes are nanoscale vesicles secreted by most cells, which can carry biologically active molecules and mediate communication and cargo transportation among cells. Recent studies have focused on transforming exosomes into good drug delivery systems (DDSs) to improve the clinical treatment of PDAC. This review considers PDAC as the main research object to introduce the role of PDAC-exosomes in PDAC development and metastasis. This review focuses on the following two themes: (a) the great potential of PDAC-exosomes as new diagnostic markers for PDAC, and (b) the transformation of exosomes into potential DDSs.


Assuntos
Carcinoma Ductal Pancreático , Exossomos , Neoplasias Pancreáticas , Humanos , Exossomos/patologia , Linhagem Celular Tumoral , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Sistemas de Liberação de Medicamentos , Neoplasias Pancreáticas
3.
Mol Ther ; 30(2): 662-671, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34400328

RESUMO

Pancreatic cancer is resistant to conventional therapeutic interventions, mainly due to abundant cancer stromal cells and poor immune cell infiltration. Here, we used a targeted cancer therapy approach based on attenuated Salmonella typhimurium engineered to express cytolysin A (ClyA) to target cancer stromal cells and cancer cells and treat pancreatic cancer in mice. Nude mice bearing subcutaneous or orthotopic human pancreatic cancers were treated with engineered S. typhimurium expressing ClyA. The tumor microenvironment was monitored to analyze stromal cell numbers, stromal cell marker expression, and immune cell infiltration. The attenuated bacteria accumulated and proliferated specifically in tumor tissues after intravenous injection. The bacteria secreted ClyA into the tumor microenvironment. A single dose of ClyA-expressing Salmonella markedly inhibited growth of pancreatic cancer both in subcutaneous xenograft- and orthotopic tumor-bearing nude mice. Histological analysis revealed a marked decrease in expression of stromal cell markers and increased immune cell (neutrophils and macrophages) infiltration into tumors after colonization by ClyA-expressing bacteria. ClyA-expressing S. typhimurium destroyed cancer stromal cells and cancer cells in mouse models of human pancreatic cancer. This approach provides a novel strategy for combining anticancer and anti-stromal therapy to treat pancreatic cancer.


Assuntos
Neoplasias Pancreáticas , Salmonella typhimurium , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Células Estromais , Microambiente Tumoral
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(2): 268-274, 2023 Feb 28.
Artigo em Inglês, Zh | MEDLINE | ID: mdl-36999474

RESUMO

Pancreatic cancer (PC) is a malignant tumor of the digestive tract with poor patient prognosis. The PC incidence is still increasing with a 5-year survival rate of only 10%. At present, surgical resection is the most effective method to treat PC, however, 80% of the patients missed the best time for surgery after they have been diagnosed as PC. Chemotherapy is one of the main treating methods but PC is insensitive to chemotherapy, prone to drug resistance, and is accompanied by many side effects which are related to a lack of specific target. Exosomes are nanoscale vesicles secreted by almost all cell types and can carry various bioactive substances which mediate cell communication and material transport. They are characterized by a low immunogenicity, low cytotoxicity, high penetration potential and homing capacity, and possess the potential of being used as advanced drug carriers. Therefore, it is a hot research topic to use drug-loaded exosomes for tumor therapy. They may alleviate chemotherapy resistance, reduce side effects, and enhance the curative effect. In recent years, exosome drug carriers have achieved considerable results in PC chemotherapy studies.


Assuntos
Antineoplásicos , Exossomos , Neoplasias Pancreáticas , Humanos , Exossomos/metabolismo , Portadores de Fármacos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/diagnóstico , Antineoplásicos/uso terapêutico
5.
BMC Surg ; 22(1): 127, 2022 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-35366868

RESUMO

BACKGROUND: To explore the application value of free omental wrapping and modified pancreaticojejunostomy in pancreaticoduodenectomy (PD). METHODS: The clinical data of 175 patients who underwent pancreaticoduodenectomy from January 2015 to December 2020 were retrospectively analysed. In total, 86 cases were divided into Group A (omental wrapping and modified pancreaticojejunostomy) and 89 cases were divided into Group B (control group). The incidences of postoperative pancreatic fistula and other complications were compared between the two groups, and univariate and multivariate logistic regression analyses were used to determine the potential risk factors for postoperative pancreatic fistula. Risk factors associated with postoperative overall survival were identified using Cox regression. RESULTS: The incidences of grade B/C pancreatic fistula, bile leakage, delayed bleeding, and reoperation in Group A were lower than those in Group B, and the differences were statistically significant (P < 0.05). Group A had an earlier drainage tube extubation time, earlier return to normal diet time and shorter postoperative hospital stay than the control group (P < 0.05). The levels of C-reactive protein (CRP), interleukin-6 (IL-6), and procalcitonin (PCT) inflammatory factors 1, 3 and 7 days after surgery also showed significant. Univariate and multivariate logistic regression analyses showed that a body mass index (BMI) ≥ 24, pancreatic duct diameter less than 3 mm, no isolation of the greater omental flap and modified pancreaticojejunostomy were independent risk factors for pancreatic fistula (P < 0.05). Cox regression analysis showed that age ≥ 65 years old, body mass index ≥ 24, pancreatic duct diameter less than 3 mm, no isolation of the greater omental flap isolation and modified pancreaticojejunostomy, and malignant postoperative pathology were independent risk factors associated with postoperative overall survival (P < 0.05). CONCLUSIONS: Wrapping and isolating the modified pancreaticojejunostomy with free greater omentum can significantly reduce the incidence of postoperative pancreatic fistula and related complications, inhibit the development of inflammation, and favourably affect prognosis.


Assuntos
Pancreaticoduodenectomia , Pancreaticojejunostomia , Idoso , Humanos , Pancreatectomia/efeitos adversos , Fístula Pancreática/epidemiologia , Fístula Pancreática/etiologia , Pancreaticoduodenectomia/efeitos adversos , Pancreaticojejunostomia/efeitos adversos , Estudos Retrospectivos
6.
World J Surg Oncol ; 19(1): 285, 2021 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-34548086

RESUMO

OBJECTIVE: To investigate the effect of Helicobacter pylori (H. pylori) eradication on the prognosis of postoperative early gastric cancer (EGC). METHODS: This is a retrospective study based on data from 6 hospitals. We identified 429 patients with EGC who underwent curative gastrectomy from January 2010 to December 2016. All of the patients were tested for H. pylori. Patients were divided into two groups, the successful H. pylori eradication group (group A, 268 patients) and the non-H. pylori eradication group (group B, 161 patients), for calculating the disease-free survival (DFS) and overall survival (OS) of each group. RESULT: Positive node metastasis (hazard ratio (HR), 3.13; 95% confidence interval (CI), 1.84-5.32; P < 0.001), undifferentiated type (HR, 2.54; 95% CI, 1.51-4.28; P < 0.001), and non-H. pylori eradication (HR, 1.73; 95% CI, 1.08-2.77; P = 0.023) were statistically significantly independent risk factors of recurrence. Patient's age ≥60 years old (HR, 3.32; 95% CI, 2.00-5.53; P < 0.001), positive node metastasis (HR, 3.71; 95% CI, 2.25-6.12; P < 0.001), undifferentiated type (HR, 3.06; 95% CI, 1.79-5.23; P < 0.001), and non-H. pylori eradication (HR, 1.83; 95% CI, 1.11-3.02; P = 0.018) were statistically significantly independent risk factors of overall survival. CONCLUSION: H. pylori eradication treatment could prevent the recurrence of postoperative EGC to prolong the overall survival of patients with EGC.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Antibacterianos/uso terapêutico , Gastroscopia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/tratamento farmacológico , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/epidemiologia , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/cirurgia
7.
Pancreatology ; 18(3): 328-333, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29525378

RESUMO

OBJECTIVE: To explore the molecular mechanisms of celecoxib-induced pancreatic cancer suppression in vivo and in vitro. METHODS: The anti-pancreatic cancer activities of celecoxib (0, 20, 60 and 100 µmol/L) were investigated by cell viability and migration of Panc-1 and Bxpc-3 cells in vitro. The expression of L1CAM in pancreatic cancer and adjacent tissues was compared using immunohistochemistry. The expressions of L1CAM, STAT3, p-STAT3, NF-κB, p-NF-κB were determined by western blotting, and cell invasive ability was determined by wound healing assay in L1CAM-silenced and over-expressed Panc-1and Bxpc-3 cells. RESULTS: The expression of L1CAM in pancreatic carcinoma was stronger than that in the adjacent tissues and L1CAM could increase the growth and invasion of pancreatic cancer cells. Over-expression of L1CAM activated the STAT3/NF-κB signaling pathway in Panc-1 and Bxpc-3 pancreatic cancer cells and celecoxib inhibited their viability and the expressions of STAT3, p-STAT3, NF-κB, p-NF-κB as well as full length L1CAM in a concentration dependent manner. CONCLUSIONS: L1CAM was highly expressed in pancreatic cancer tissue and positively correlated with age, TNM staging and tumor differentiation. L1CAM activated the STAT/NF-κB signaling pathway and celecoxib could inhibit the activity of L1CAM, STAT3 and the NF-κB signaling pathway resulting in decreased growth and invasion of pancreatic cancer cells.


Assuntos
Antígeno CD56/antagonistas & inibidores , Celecoxib/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Metástase Neoplásica/prevenção & controle , Neoplasias Pancreáticas/prevenção & controle , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição RelA/antagonistas & inibidores , Antígeno CD56/genética , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Imuno-Histoquímica , Invasividade Neoplásica/genética , Neoplasias Pancreáticas/patologia , Plasmídeos/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Cicatrização/efeitos dos fármacos , Cicatrização/genética
8.
Mol Ther ; 25(10): 2299-2308, 2017 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-28865999

RESUMO

Hepatocellular carcinoma (HCC) has a high morbidity and mortality rate worldwide, with limited treatment options. Glypican-3 (GPC3) is a glycosylphosphatidylinositol-anchored glycoprotein that is overexpressed in most HCC tissues but not in normal tissues. GPC3-targeting antibody therapy shows limited response in a clinical trial due to the lack of a tumor-specific cytotoxic T lymphocyte (CTL) response. Here, in C57/B6 mice, we demonstrated that intravenous infusion of GPC3-coupled lymphocytes (LC/GPC3+) elicited robust GPC3-specific antibody and CTL responses, which effectively restricted proliferation and lysed cultured-HCC cells. Treatment with LC/GPC3+ induced durable tumor regression in HCC-bearing C57/B6 mice. Administration of LC/GPC3+ induced elevated levels of the cytotoxic T cell bioactive factors tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ), granzyme B, and perforin, and substantially increased the number of infiltrating CD8+ T cells in tumor tissues. Moreover, immune responses elicited by LC/GPC3+ selectively suppressed GPC3+ tumors, but didn't affect the GPC3- tumors in BALB/c mice. Our findings provide the first preclinical evidence that intravenous infusion of the LC/GPC3+ complex can induce a strong anti-HCC effect through regulating systemic and local immune responses. These results indicate that the LC/GPC3+ complex could be developed as precision therapeutics for HCC patients in the future.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/prevenção & controle , Glipicanas/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Interferon gama/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
9.
Tumour Biol ; 39(6): 1010428317710410, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28639887

RESUMO

Hepatocellular carcinoma is a leading cause of cancer-related mortality worldwide. TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) is a potential target for cancer therapy. However, many cancer cells are resistant to TRAIL-induced apoptosis and its mechanism is not well understood. In this study, to identify potential therapeutic targets for TRAIL-resistant cancer cells, we compared the expression levels of interferon-stimulated gene 15 in TRAIL-sensitive and TRAIL-resistant hepatocellular carcinoma cell lines. Western blot analysis showed that interferon-stimulated gene 15 expression levels were significantly higher in resistant HLCZ01and Huh7 cells than in sensitive LH86 and SMMC-7721 cells. Interferon-stimulated gene 15 knockdown in resistance cells led to TRAIL sensitivity. Conversely, interferon-stimulated gene 15 overexpression in sensitive cells resulted in TRAIL resistance. Our bioinformatics search detected a putative target sequence for microRNA miR-138 in the 3' untranslated region of the interferon-stimulated gene 15. Real-time quantitative polymerase chain reaction analysis demonstrated that miR-138 was significantly downregulated in TRAIL-resistant cells compared to TRAIL-sensitive cells. Forced expression of miR-138 in resistant cells decreased both messenger RNA and protein levels of interferon-stimulated gene 15, and when exposed to TRAIL, activated poly(adenosine diphosphate-ribose) polymerase, indicating sensitization to TRAIL. The results suggested that miR-138 regulates the interferon-stimulated gene 15 expression by directly targeting the 3' untranslated region of interferon-stimulated gene 15 and modulates the sensitivity to TRAIL-induced apoptosis. MiR-138 may be a target for therapeutic intervention in TRAIL-based drug treatments of resistant hepatocellular carcinoma or could be a biomarker to select patients who may benefit from the treatment.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Regiões 3' não Traduzidas/genética , Apoptose/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Interferons/genética , Neoplasias Hepáticas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(13): E1264-73, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24616513

RESUMO

The absence of a robust cell culture system for hepatitis B virus (HBV) and hepatitis C virus (HCV) infection has limited the analysis of the virus lifecycle and drug discovery. We have established a hepatoma cell line, HLCZ01, the first cell line, to the authors' knowledge, supporting the entire lifecycle of both HBV and HCV. HBV surface antigen (HBsAg)-positive particles can be observed in the supernatant and the lumen of the endoplasmic reticulum of the cells via electron microscopy. Interestingly, HBV and HCV clinical isolates propagate in HLCZ01 cells. Both viruses replicate in the cells without evidence of overt interference. HBV and HCV entry are blocked by antibodies against HBsAg and human CD81, respectively, and the replication of HBV and HCV is inhibited by antivirals. HLCZ01 cells mount an innate immune response to virus infection. The cell line provides a powerful tool for exploring the mechanisms of virus entry and replication and the interaction between host and virus, facilitating the development of novel antiviral agents and vaccines.


Assuntos
Carcinoma Hepatocelular/virologia , Hepacivirus/fisiologia , Vírus da Hepatite B/fisiologia , Neoplasias Hepáticas/virologia , Replicação Viral/fisiologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Coinfecção/tratamento farmacológico , Coinfecção/patologia , Coinfecção/virologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/crescimento & desenvolvimento , Hepatite B/tratamento farmacológico , Hepatite B/virologia , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/crescimento & desenvolvimento , Vírus da Hepatite B/isolamento & purificação , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Replicação Viral/efeitos dos fármacos
11.
Exp Cell Res ; 333(2): 316-326, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25724899

RESUMO

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Interferon-alpha (IFN-α) has recently been recognized to harbor therapeutic potential in the prevention and treatment of HCC, but it remains controversial as to whether IFN-α exerts direct cytotoxicity against HCC. Cyclooxygenase-2 (COX-2) is overexpressed in HCC and is considered to play a role in hepatocarcinogenesis. Therefore, we aimed to elucidate the combined effect of a COX-2 inhibitor, celecoxib, and IFN-α on in vitro growth suppression of HCC using the hepatoma cell line HLCZ01 and the in vivo nude mouse xenotransplantation model using HLCZ01 cells. Treatment with celecoxib and IFN-α synergistically inhibited cell proliferation in a dose- and time-dependent manner. Apoptosis was identified by 4׳,6-diamidino-2-phenylindole dihydrochloride and fluorescent staining. IFN-α upregulated the expression of TRAIL, while celecoxib increased the expression of TRAIL receptors. The combined regimen with celecoxib and IFN-α reduced the growth of xenotransplanted HCCs in nude mice. The regulation of IFN-α- and COX-2 inhibitor-induced cell death is impaired in a subset of TRAIL-resistant cells. The molecular mechanisms of HCC cells resistant to TRAIL-induced apoptosis were explored using molecular biological and immunological methods. Interferon-α and the COX-2 inhibitor celecoxib synergistically increased TRAIL-induced apoptosis in hepatocellular carcinoma. These data suggest that IFN-α and celecoxib may offer a novel role with important implications in designing new therapeutics for TRAIL-resistant tumors.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Interferon-alfa/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Carcinoma Hepatocelular/patologia , Caspases/metabolismo , Celecoxib , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase 2/farmacologia , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Zhonghua Yi Xue Za Zhi ; 94(42): 3310-3, 2014 Nov 18.
Artigo em Zh | MEDLINE | ID: mdl-25622629

RESUMO

OBJECTIVE: To explore the major risk factors for pulmonary infection after radical gastrectomy in patients with gastric cancer. METHODS: From November 2010 to February 2014, a total of 765 patients undergoing radical gastrectomy at our hospital were divided into 2 groups based upon the presence of postoperative pulmonary infection (n = 32, 4.2%) or not (n = 733, 95.8%). Their clinicopathological data were retrospectively analyzed by Logistic regressive analysis with a case-control study model. RESULTS: Comparing with the control group, the patients had longer surgical duration (245.7 ± 66.7 vs 210.9 ± 47.2 min, P < 0.01), higher rates of requiring intensive care (12.50% vs 2.86%, P = 0.02) and longer post-operative hospital stays (21.9 ± 24.9 vs 14.2 ± 4.2 days, P < 0.01) in the postoperative pulmonary infection group.Univariate Logistic regressive analysis found that age ≥ 60 years, smoking ≥ 400 year by cigarette, diabetes mellitus, chronic obstructive pulmonary disease, proximal or total gastrectomy, combined organ resection, surgical duration ≥ 240 min, intra-operative blood loss ≥ 300 ml, peri-operative transfusion, transfusion ≥ 3 unit packed red blood cell, post-operative transfusion and post-operative complications other than pulmonary infections were associated with postoperative pulmonary infection (all P < 0.05).Further multivariate analysis identified 4 independent risk factors for pulmonary infection after radical gastrectomy, including diabetes mellitus (OR = 4.77, 95%CI:1.18-19.23), post-operative complications other than pulmonary infections (OR = 3.15, 95%CI:1.25-7.90), intra-operative blood loss ≥ 300 ml (OR = 2.63, 95%CI:1.17-5.90) and post-operative nasogastric tube ≥ 5 days (OR = 2.30, 95%CI:1.02-5.21). CONCLUSION: Correcting the modifiable risk factors may reduce the incidence of pulmonary infection and shorten the length of hospital stays and costs after radical gastrectomy in patients with gastric cancer.


Assuntos
Gastrectomia , Pneumonia , Complicações Pós-Operatórias , Neoplasias Gástricas , Transfusão de Sangue , Estudos de Casos e Controles , Doenças Transmissíveis , Humanos , Pessoa de Meia-Idade , Análise Multivariada , Transfusão de Plaquetas , Doença Pulmonar Obstrutiva Crônica , Estudos Retrospectivos , Fatores de Risco , Fumar
13.
Zhonghua Yi Xue Za Zhi ; 94(10): 751-4, 2014 Mar 18.
Artigo em Zh | MEDLINE | ID: mdl-24844958

RESUMO

OBJECTIVE: To explore the association of perioperative homologous blood transfusion (packed red blood cell, PRBC) and postoperative complications after radical gastrectomy in patients with gastric cancer. METHODS: From October 2010 to July 2013, a total of 636 patients undergoing radical gastrectomy at Department of Gastric, Duodenal & Pancreatic Surgery at Hunan Provincial Tumor Hospital were divided into 2 groups according to perioperative blood transfusion (n = 170, 26.73%) or not (n = 466, 73.27%). Their clinicopathological data, such as age, gender, co-morbidities, surgical duration, intraoperative blood loss volume and pathological stage were retrospectively analyzed by case-control study model. And the transfusion group was further divided into subgroup by transfusion volume (total PRBC<3.0, 3.0-7.5 or >7.5 U) and timing (pre-, intra- or post-operative) to examine the association of transfusion volume and timing with postoperative complications by Logistic regression. RESULTS: Thirty-two patients suffered from complications in the transfusion group (18.82%). And it was significantly more common than that in the control group (10.09% (47/466) , P < 0.01). Moreover, the complication rate (33.33% (12/36) ) was obviously higher in the large transfusion volume group (PRBC>7.5 U) than with those in the moderate (15.53% (16/103), P = 0.02) and low groups (12.90% (4/31) , P = 0.04). Infection was more common along with the total amount of transfused blood (6.45% (2/31), 10.68% (11/103) and 19.44% (7/36) in the low, moderate and large transfusion group respectively). Yet the differences were insignificant (P = 0.22). There was no significant difference of complication rates among the pre-, intra- and post-operative transfusion group classified by transfusion time (P = 0.39). And the postoperative infection rates were also insignificantly different (P = 0.88). Further Logistic analysis revealed that perioperative transfusion (OR = 2.71, 95% CI: 1.40-5.27, P < 0.01) was an independent risk factor for postoperative complications after radical gastrectomy. CONCLUSIONS: Perioperative blood transfusion is significantly associated with postoperative complications after radical gastrectomy in patients with gastric cancer. And a positive correlation exists between infection and the amount of transfused blood. But there was no association between transfusion time and complications. Thus decreasing perioperative transfusion may reduce the incidence of postoperative complications and shorten the length of hospital stays.


Assuntos
Complicações Pós-Operatórias/etiologia , Neoplasias Gástricas/cirurgia , Reação Transfusional , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Gastrectomia/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Período Perioperatório , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Adulto Jovem
14.
iScience ; 27(4): 109533, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38591006

RESUMO

Hepatitis B virus (HBV) infection generally elicits weak type-I interferon (IFN) immune response in hepatocytes, covering the regulatory effect of IFN-stimulated genes. In this study, low level of IFN-stimulated gene 12a (ISG12a) predicted malignant transformation and poor prognosis of HBV-associated hepatocellular carcinoma (HCC), whereas high level of ISG12a indicated active NK cell phenotypes. ISG12a interacts with TRIM21 to inhibit the phosphorylation activation of protein kinase B (PKB, also known as AKT) and ß-catenin, suppressing PD-L1 expression to block PD-1/PD-L1 signaling, thereby enhancing the anticancer effect of NK cells. The suppression of PD-1-deficient NK-92 cells on HBV-associated tumors was independent of ISG12a expression, whereas the anticancer effect of PD-1-expressed NK-92 cells on HBV-associated tumors was enhanced by ISG12a and treatments of atezolizumab and nivolumab. Thus, tumor intrinsic ISG12a promotes the anticancer effect of NK cells by regulating PD-1/PD-L1 signaling, presenting the significant role of innate immunity in defending against HBV-associated HCC.

15.
Carcinogenesis ; 34(5): 1001-5, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23299404

RESUMO

Germline mutations in genes that cause hereditary syndromes are highly predisposed to familial pancreatic cancer. However, genetic susceptibility to sporadic pancreatic cancer is largely uncovered. We conducted a two-stage association study on pancreatic cancer that included 981 cases and 1991 controls in the first stage followed by a second stage (2603 cases and 2877 controls). Using an approach based on candidate genes whose roles in pancreatic cancer have been well known, we identified two new susceptibility loci. rs11571836 located in the BRCA2 3'-untranslated region was significantly associated with lower expression of BRCA2 transcript and increased pancreatic cancer risk [odds ratio = 1.30, 95% confidence interval = 1.14-1.47, P = 7.64 × 10(-5)] in a recessive manner. rs12939944 located in the MAP2K4 intron was associated with decreased risk (odds ratio = 0.82, 95% confidence interval = 0.74-0.91, P = 0.0001) in a dominant manner. Our results demonstrate for the first time that common variants in BRCA2 and MAP2K4 are susceptibility to sporadic pancreatic cancer.


Assuntos
Proteína BRCA2/genética , MAP Quinase Quinase 4/genética , Neoplasias Pancreáticas/genética , Regiões 3' não Traduzidas , Estudos de Casos e Controles , Predisposição Genética para Doença , Genótipo , Mutação em Linhagem Germinativa , Humanos , Íntrons , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/metabolismo , Polimorfismo de Nucleotídeo Único
16.
Zhonghua Yi Xue Za Zhi ; 93(40): 3211-4, 2013 Oct 29.
Artigo em Zh | MEDLINE | ID: mdl-24405543

RESUMO

OBJECTIVE: To explore the major risk factors for intra-abdominal infections after radical gastrectomy in patients with gastric cancer. METHODS: From October 2010 to January 2013, a total of 479 patients undergoing radical gastrectomy at Department of Gastric, Duodenal & Pancreatic Surgery, Hunan Provincial Tumor Hospital were divided into 2 groups according to an onset of postoperative intra-abdominal infections (n = 32, 6.68%) or not (n = 447, 93.32%). Their clinicopathological data, such as age, gender, co-morbidities, surgical duration, operative blood loss and pathological stage were retrospectively analyzed by Logistic regressive analysis with a case-control study model. RESULTS: As compared with the control group, the patients had a greater age ((59 ± 10) vs (53 ± 11) years, P < 0.01), lower lymphocyte count ((1.4 ± 0.7) ×10(9)/L vs (1.7 ± 0.6) ×10(9)/L, P = 0.02), lower hemoglobin level ( (108 ± 28) vs (117 ± 24) g/L, P = 0.04), lower albumin level ((34 ± 6) vs (37 ± 5) g/L, P < 0.01) and longer surgical duration ((244 ± 43) vs (216 ± 45) min, P < 0.01) in the postoperative intra-abdominal infection group. Univariate Logistic regressive analysis found that a history of abdominal surgery, body mass index (BMI) >25 kg/m(2), co-morbidities, diabetes mellitus, complications due to gastric cancer, lymphocyte count <1.5×10(9)/L, hemoglobin <100 g/L, albumin <30 g/L, ascites, perioperative transfusion, total mastectomy, combined organ resection and surgical duration >240 min were associated with the occurrence of postoperative intra-abdominal infections (all P < 0.05). Further multivariate analysis identified 4 independent risk factors for intra-abdominal infections after radical gastrectomy, including combined multiorgan resection (OR = 3.64, 95%CI: 1.39-9.55), BMI>25 kg/m(2) (OR = 3.04, 95%CI: 1.17-7.92), diabetes mellitus (OR = 3.41, 95%CI: 1.05-11.09) and perioperative transfusion (OR = 2.24, 95%CI: 1.02-5.13). CONCLUSION: A correction of modifiable risk factors may reduce the incidence of intra-abdominal infections after radical gastrectomy, shorten the length of hospital stays and improve outcomes in patients with gastric cancer.


Assuntos
Gastrectomia/efeitos adversos , Infecções Intra-Abdominais/etiologia , Neoplasias Gástricas/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fatores de Risco , Neoplasias Gástricas/cirurgia , Adulto Jovem
17.
Zhonghua Yi Xue Za Zhi ; 93(46): 3667-70, 2013 Dec.
Artigo em Zh | MEDLINE | ID: mdl-24534346

RESUMO

OBJECTIVE: To explore the complications after radical gastrectomy in patients with gastric cancer according to Clavien-Dindo classification and examine the major risk factors for complications. METHODS: From October 2010 to June 2013, a total of 614 patients undergoing radical gastrectomy at Department of Gastric,Duodenal & Pancreatic Surgery at Hunan Provincial Tumor Hospital were divided into 2 groups according to the occurrence of complications (n = 76, 12.38%) or not (n = 538, 87.62%). Their clinicopathological data, such as age, gender, co-morbidities, surgical duration, operative blood loss volume and pathological stage were retrospectively analyzed by Logistic regression with a case-control model. RESULTS: Among them, 76 patients developed complications (12.38%). According to Clavien-Dindo classification, 56(9.12%), 14(2.28%), 3(0.49%) and 3(0.49%) patients suffered stage II, III, IV and V complications respectively. Comparing with the control group, the patients had a higher transfusion rate (43.42% (n = 33) vs 24.16% (n = 130), P < 0.01) and a longer postoperative hospital stay in the complication group ((23 ± 18) vs (14 ± 6) days, P < 0.01). There was no difference in age, gender, body mass index (BMI), number of dissected lymph node, levels of hemoglobin and albumin at admission, intraoperative hemorrhage, surgical duration and pathological TNM stage in two groups (all P > 0.05). Univariate analysis revealed that BMI > 25 kg/m(2), co-morbidities, diabetes mellitus, complications due to gastric cancer, hemoglobin <100 g/L, albumin <30 g/L, ascites, total gastrectomy, combined multi-organ resection, surgical duration >240 min and perioperative transfusion were associated with postoperative complications (all P < 0.05).Further multivariate analysis showed that perioperative transfusion (OR = 2.78, 95%CI: 1.42-5.43, P < 0.01) and combined multi-organ resection (OR = 1.72, 95%CI: 1.14-2.58, P = 0.01) were independent risk factors for postoperative complications after radical gastrectomy. CONCLUSIONS: Classifying the complications after radical gastrectomy according to Clavien-Dindo classification is important for comparisons and quality assessments among different studies. And decreasing perioperative transfusion and avoiding combined multi-organ resection may reduce the incidence of postoperative complications and shorten the length of hospital stay.


Assuntos
Gastrectomia/efeitos adversos , Complicações Pós-Operatórias/patologia , Neoplasias Gástricas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Complicações Pós-Operatórias/epidemiologia , Estudos Retrospectivos , Fatores de Risco , Adulto Jovem
18.
Oncol Rep ; 49(5)2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36928140

RESUMO

Gastric diffuse large B­cell lymphoma (GDLBCL) is a common disease with an increasing incidence. However, the regulatory effect of exosomal programmed death­ligand 1 (PD­L1) on the immune microenvironment in GDLBCL is unclear. In the present study, the protein expression levels of exosomal PD­L1 in the supernatants of cultured diffuse large B­cell lymphoma (DLBCL) cells and the plasma of patients with GDLBCL was assessed using immunoblotting. Exosomes derived from DLBCL cells were cocultured with T lymphocytes or injected into tumor xenograft mice by tail vein injection. The relationship between the protein expression level of exosomal PD­L1 in the plasma and the clinical characteristics and immune microenvironmental parameters of GDLBCL was evaluated using immunoblotting and immunohistochemistry. High levels of exosomal PD­L1 were found in the supernatants of cultured DLBCL cells. Exosomes with high levels of PD­L1 promoted growth of tumors formed by DLBCL cells in vivo and inhibited the proliferation of T lymphocytes. Notably, the protein expression level of PD­L1 in plasma exosomes derived from GDLBCL patients was significantly higher than that of healthy individuals. High levels of PD­L1 in plasma exosomes were significantly associated with international prognostic index score, pathological type and advanced Lugano stage, which might lead to the poor prognosis of GDLBCL. Moreover, a high level of PD­L1 in plasma exosomes was significantly associated with an immunosuppressive microenvironment in GDLBCL. Therefore, the results of the present study indicated that exosomal PD­L1 inhibited the proliferation of T lymphocytes and promoted the formation of an immunosuppressive microenvironment in GDLBCL. High expression of exosomal PD­L1 may suggest a poor prognosis of GDLBCL, and exosomal PD­L1 in plasma may be a new diagnostic indicator for GDLBCL.


Assuntos
Exossomos , Linfoma Difuso de Grandes Células B , Neoplasias Gástricas , Microambiente Tumoral , Animais , Humanos , Camundongos , Antígeno B7-H1/metabolismo , Exossomos/metabolismo , Imunossupressores/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Neoplasias Gástricas/metabolismo , Microambiente Tumoral/imunologia
19.
iScience ; 26(12): 108414, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38047083

RESUMO

Cytotoxin-associated gene A (CagA) of Helicobacter pylori (Hp) may promote immune evasion of Hp-infected gastric cancer (GC), but potential mechanisms are still under explored. In this study, the positive rates of CagA and PD-L1 protein in tumor tissues and the high level of exosomal PD-L1 protein in plasma exosomes were significantly associated with the elevated stages of tumor node metastasis (TNM) in Hp-infected GC. Moreover, the positive rate of CagA was positively correlated with the positive rate of PD-L1 in tumor tissues and the level of PD-L1 protein in plasma exosomes, and high level of exosomal PD-L1 might indicate poor prognosis of Hp-infected GC. Mechanically, CagA increased PD-L1 level in exosomes derived from GC cells by inhibiting p53 and miRNA-34a, suppressing proliferation and anticancer effect of CD8+ T cells. This study provides sights for understanding immune evasion mediated by PD-L1. Targeting CagA and exosomal PD-L1 may improve immunotherapy efficacy of Hp-infected GC.

20.
Oncol Lett ; 23(5): 153, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35836485

RESUMO

A previous bioinformatics study suggested that circular RNA 0001666 (circ_0001666) and its target microRNA (miR)-1229 were associated with colorectal cancer (CRC) pathogenesis. However, the role of this interaction in the regulation of CRC cell malignancy remains unclear. Thus, the aim of the present study was to examine the interaction between circ_0001666 and miR-1229, and its effects on CRC cell malignancy. circ_0001666 overexpression or knockdown plasmids were transfected into the HT-29 and HCT-116 cell lines. In addition, in rescue experiments, circ_000166 or miR-1229 overexpression plasmids were transfected into the HT-29 cell line, either alone or in combination. Following transfection, cell proliferation, apoptosis, invasion and the number of CD133+ cells were analyzed. The protein expression level of proteins in the Wnt/ß-catenin pathway was also examined. In both HT-29 and HCT-116 cell lines, circ_0001666 overexpression increased apoptosis, whilst inhibiting cell proliferation and invasion, and reducing the frequency of CD133+ cells. By contrast, circ_0001666 knockdown reduced apoptosis, but increased cell proliferation and the number of CD133+ cells. However, cell invasion remained unaffected. In addition, circ_0001666 expression levels negatively regulated those of miR-1229, whereas miR-1229 expression did not affect circ_0001666, in both the HT-29 and HCT-116 cell lines. Furthermore, a luciferase reporter assay confirmed that miR-1229 directly bound to circ_0001666. In the HT-29 cell line, miR-1229 overexpression activated the Wnt/ß-catenin pathway, and promoted cell proliferation, invasion and stemness, while suppressing cell apoptosis. In addition, miR-1229 overexpression reversed the effects of circ_0001666 overexpression. In conclusion, circ_0001666 suppresses CRC cell proliferation, invasion and stemness by inhibiting the Wnt/ß-catenin signaling pathway by targeting miR-1229, and may represent a potential target for CRC treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA