Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
7.
BMC Cancer ; 23(1): 1076, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37936091

ABSTRACT

BACKGROUND: The clinical profiles of recurrent retroperitoneal liposarcoma (RLS) need to be explored. The recurrence patterns of RLS are controversial and ambiguous. METHODS: A total of 138 patients with recurrent RLS were finally recruited in the study. The analysis of overall survival (OS) and recurrence-free survival (RFS) was performed by Kaplan‒Meier analysis. To identify independent prognostic factors, all significant variables on univariate Cox regression analysis (P ≤ 0.05) were subjected to multivariate Cox regression analysis. The corresponding nomogram model was further built to predict the survival status of patients. RESULTS: Among patients, the 1-, 3-, and 5-year OS rates were 70.7%, 35.9% and 30.9%, respectively. The 1-, 3- and 5-year RFS rates of the 55 patients who underwent R0 resection were 76.1%, 50.8% and 34.4%, respectively. The multivariate analysis revealed that resection method, tumor size, status of pathological differentiation, pathological subtypes and recurrence pattern were independent risk factors for OS or RFS. Patients with distant recurrence (DR) pattern usually had multifocal tumors (90.5% vs. 74.7%, P < 0.05); they were prone to experience changes of pathological differentiation (69.9% vs. 33.3%, P < 0.05) and had a better prognosis than those with local recurrence (LR) pattern. R0 resection and combined organ resection favored the survival of patients with DR pattern in some cases. CONCLUSIONS: Patients with DR pattern had better prognosis, and they may benefit more from aggressive combined resection than those with LR pattern. Classifying the recurrence patterns of RLS provides guidance for individualized clinical management of recurrent RLS.


Subject(s)
Liposarcoma , Retroperitoneal Neoplasms , Humans , Neoplasm Recurrence, Local/pathology , Liposarcoma/pathology , Retroperitoneal Neoplasms/surgery , Retroperitoneal Neoplasms/pathology , Retroperitoneal Space/pathology , Prognosis , Survival Rate , Retrospective Studies
8.
Mol Nutr Food Res ; 67(24): e2300376, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37815169

ABSTRACT

SCOPE: Ulcerative colitis (UC) is an intestinal disease that is becoming increasingly prevalent and is often overlooked in early stages, and its pathogenesis is often closely related to inflammatory processes. Betaine is a natural product with anti-inflammatory effects that exists in a wide range of plants and animals. METHODS AND RESULTS: In this study, the protective effects of betaine are investigated on intestinal barrier function in a mouse model, a dextran sulfate sodium-induced ulcerative colitis and its mechanism of action in the inflammatory context. FITC-dextran 4000 Da (FD-4) flux, disease activity index, histopathological scores, and inflammatory factor levels in sera are determined across different groups. In addition, Caco-2 cell monolayer barrier function is evaluated by transepithelial resistance and FD-4 flux. The expression levels and distribution of tight junction proteins are determined using Western blot and immunofluorescence, respectively. Activation of the NF-κBp65/MLCK/p-MLC signaling pathway is detected by Western blot. Chromatin immunoprecipitation is performed to examine the binding of NF-κB to the MLCK gene promoter. The results indicated that betaine inhibits NF-κB-mediated activation of the MLCK/p-MLC signaling pathway to protect the intestinal barrier function of mice with UC. CONCLUSION: Betaine can be used as a potential candidate drug to improve intestinal barrier dysfunction in patients with UC.


Subject(s)
Colitis, Ulcerative , Colitis , Humans , Mice , Animals , NF-kappa B/metabolism , Colitis, Ulcerative/chemically induced , Caco-2 Cells , Dextran Sulfate/toxicity , Betaine/pharmacology , Betaine/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Intestinal Mucosa/metabolism
10.
Biochem Pharmacol ; 216: 115752, 2023 10.
Article in English | MEDLINE | ID: mdl-37634598

ABSTRACT

This study aimed to investigate the protective effects of S-adenosylmethionine (SAM) on irinotecan-induced intestinal barrier dysfunction and microbial ecological dysregulation in both mice and human colon cell line Caco-2, which is widely used for studying intestinal epithelial barrier function. Specifically, this study utilized Caco-2 monolayers incubated with 7-ethyl-10-hydroxycamptothecin (SN-38) as well as an irinotecan-induced diarrhea model in mice. Our study found that SAM pretreatment significantly reduced body weight loss and diarrhea induced by irinotecan in mice. Furthermore, SAM inhibited the increase of intestinal permeability in irinotecan-treated mice and ameliorated the decrease of Zonula occludens-1(ZO-1), Occludin, and Claudin-1 expression. Additionally, irinotecan treatment increased the relative abundance of Proteobacteria compared to the control group, an effect that was reversed by SAM administration. In Caco-2 monolayers, SAM reduced the expression of reactive oxygen species (ROS) and ameliorated the decrease in transepithelial electrical resistance (TER) and increase in fluorescein isothiocyanate-dextran 4000 Da (FD-4) flux caused by SN-38. Moreover, SAM attenuated changes in the localization and distribution of ZO-1and Occludin in Caco-2 monolayers induced by SN-38 and protected barrier function by inhibiting activation of the p38 MAPK/p65 NF-κB/MLCK/MLC signaling pathway. These findings provide preliminary evidence for the potential use of SAM in treating diarrhea caused by irinotecan.


Subject(s)
Gastrointestinal Diseases , Intestinal Diseases , Humans , Animals , Mice , Irinotecan/pharmacology , Caco-2 Cells , Occludin/metabolism , Occludin/pharmacology , S-Adenosylmethionine/pharmacology , S-Adenosylmethionine/metabolism , Intestinal Mucosa , Intestinal Diseases/metabolism , Diarrhea/chemically induced , Diarrhea/drug therapy , Diarrhea/prevention & control , Tight Junctions
11.
J Hepatol ; 79(5): e188-e189, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37172649
12.
Cancer Biol Med ; 20(6)2023 05 13.
Article in English | MEDLINE | ID: mdl-37184030

ABSTRACT

OBJECTIVE: Liver cancer is a deadly malignancy associated with high mortality and morbidity. Less than 20% of patients with advanced liver cancer respond to a single anti-PD-1 treatment. The high heterogeneity of neutrophils in the tumor immune microenvironment in liver cancer may contribute to resistance to immune checkpoint blockade (ICB). However, the underlying mechanism remains largely unknown. METHODS: We established an orthotopic liver cancer model by using transposable elements to integrate the oncogenes Myc and KrasG12D into the genome in liver cells from conditional Trp53 null/null mice (pTMK/Trp53-/-). Flow cytometry and immunohistochemistry were used to assess the changes in immune cells in the tumor microenvironment. An ex vivo coculture assay was performed to test the inhibitory effects of tumor-associated neutrophils (TANs) on CD8+ T cells. The roles of neutrophils, T cells, and NK cells were validated through antibody-mediated depletion. The efficacy of the combination of neutrophil depletion and ICB was evaluated. RESULTS: Orthotropic pTMK/Trp53-/- mouse liver tumors displayed a moderate response to anti-Ly6G treatment but not PD-1 blockade. Depletion of neutrophils increased the infiltration of CD8+ T cells and decreased the number of exhausted T cells in the tumor microenvironment. Furthermore, depletion of either CD8+ T or NK cells abrogated the antitumor efficacy of anti-Ly6G treatment. Moreover, the combination of anti-Ly6G with anti-PD-L1 enhanced the infiltration of cytotoxic CD8+ T cells and thereafter resulted in a significantly greater decrease in tumor burden. CONCLUSIONS: Our data suggest that TANs may contribute to the resistance of liver cancer to ICB, and combining TAN depletion with T cell immunotherapy synergistically increases antitumor efficacy.


Subject(s)
Immune Checkpoint Inhibitors , Liver Neoplasms , Humans , Mice , Animals , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Neutrophils , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , T-Lymphocytes, Cytotoxic/pathology , Tumor Microenvironment
13.
Front Surg ; 10: 1095924, 2023.
Article in English | MEDLINE | ID: mdl-36969752

ABSTRACT

Introduction: To investigate the correlations between the Ki-67 index and plain-scan computerized tomography (CT) signs and pathological features of gastrointestinal stromal tumor (GIST) tissue. Materials and methods: Data from 186 patients with GIST diagnosed by pathology and immunohistochemistry (IHC) in Peking University First Hospital from May 2016 to May 2022 were analyzed. The patients were divided into two groups: Ki-67 ≤5% and >5%. Correlation analysis, univariate and multivariate Logistic regression analysis were used to explore the correlations between CT signs, pathological features, and Ki-67 expression. Results: Univariate indicators correlated with the Ki-67 index were mitotic count, pathological grade, tumor hemorrhage, tumor necrosis, tumor size, and tumor density. Multivariate Logistic regression indicated that the mitotic count [odds ratio (OR) 10.222, 95% confidence interval (CI) 4.312-31.039], pathological grade (OR 2.139, 95% CI 1.397-3.350), and tumor size (OR 1.096, 95% CI 1.020-1.190) were independently associated with the Ki-67 expression level. The concordance indexes (C-index) for the pathological features and CT signs models were 0.876 (95% CI 0.822-0.929) and 0.697 (95% CI 0.620-0.774), respectively, with positive predictive values of 93.62% and 58.11% and negative predictive values of 81.29% and 75.89%, respectively. After internal verification by the Bootstrap method, the fitting degree of the pathological features model was found to be better than that of the CT signs model. Conclusion: Mitotic count, pathological risk grading, and tumor size are independent risk factors correlating with high Ki-67 index. These results indicate that the Ki-67 index reflects tumor malignancy and can predict recurrence and metastasis of GIST.

14.
J Adv Res ; 44: 201-212, 2023 02.
Article in English | MEDLINE | ID: mdl-36725190

ABSTRACT

INTRODUCTION: Lipopolysaccharide (LPS) causes lesions of the epithelial barrier, which allows translocation of pathogens from the intestinal lumen to the host's circulation. Hydrogen sulfide (H2S) regulates multiple physiological and pathological processes in colonic epithelial tissue, and CBS-H2S axis involved in multiple gastrointestinal disorder. However, the mechanism underlying the effect of the CBS-H2S axis on the intestinal and systemic inflammation in colitis remains to be illustrated. OBJECTIVES: To investigate the effect of CBS-H2S axis on the intestinal and systematic inflammation related injuries in LPS induced colitis and the underlying mechanisms. METHODS: Wild type and CBS-/+ mice were used to evaluate the effect of endogenous and exogenous H2S on LPS-induced colitis in vivo. Cytokine quantitative antibody array, western blot and real-time PCR were applied to detect the key cytokines in the mechanism of action. Biotin switch of S-sulfhydration, CRISPR/Cas9 mediated knockout, immunofluorescence and ActD chase assay were used in the in vitro experiment to further clarify the molecular mechanisms. RESULTS: H2S significantly alleviated the symptoms of LPS-induced colitis in vivo and attenuated the increase of COX-2 expression. The sulfhydrated HuR increased when CBS express normally or GYY4137 was administered. While after knocking kown CBS, the expression of COX-2 in mice colon increased significantly, and the sulfhydration level of HuR decreased. The results in vitro illustrated that HuR can increase the stability of COX-2 mRNA, and the decrease of COX-2 were due to increased sulfhydration of HuR rather than the reduction of total HuR levels. CONCLUSION: These results indicated that CBS-H2S axis played an important role in protecting intestinal barrier function in colitis. CBS-H2S axis increases the sulfhydration level of HuR, by which reduces the binding of HuR with COX-2 mRNA and inhibited the expression of COX-2.


Subject(s)
Colitis , Hydrogen Sulfide , Humans , Mice , Animals , Cyclooxygenase 2 , Lipopolysaccharides , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Inflammation
15.
J Exp Clin Cancer Res ; 42(1): 44, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36755269

ABSTRACT

BACKGROUND: Advanced gastric cancer (GC) is a lethal malignancy, harboring recurrent alterations in cell cycle pathway, especially the CDKN2A-CDK4/CDK6/CCND1 axis. However, monotherapy of CDK4/6 inhibitors has shown limited antitumor effects for GC, and combination treatments were urgently needed for CDK4/6 inhibitors. METHODS: Here, we performed a comprehensive analysis, including drug screening, pan-cancer genomic dependency analysis, and epigenetic sequencing to identify the candidate combination with CDK4/6 inhibitors. Mechanisms were investigated by bulk RNA-sequencing and experimental validation was conducted on diverse in vitro or in vivo preclinical GC models. RESULTS: We found that the BRD4 inhibitor JQ1 augments the antitumor efficacy of the CDK4/6 inhibitor abemaciclib (ABE). Diverse in vitro and in vivo preclinical GC models are examined and synergistic benefits from the combination therapy are obtained consistently. Mechanistically, the combination of ABE and JQ1 enhances the cell cycle arrest of GC cells and induces unique characteristics of cellular senescence through the induction of DNA damage, which is revealed by transcriptomic profiling and further validated by substantial in vitro and in vivo GC models. CONCLUSION: This study thus proposes a candidate combination therapy of ABE and JQ1 to improve the therapeutic efficacy and worth further investigation in clinical trials for GC.


Subject(s)
Carcinoma , Stomach Neoplasms , Humans , Nuclear Proteins/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays , Transcription Factors/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics , Cell Cycle Proteins
16.
World J Gastrointest Oncol ; 14(11): 2295-2301, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36438705

ABSTRACT

BACKGROUND: Adenocarcinoma has the highest incidence among malignant tumors of the small intestine (SI). Squamous cell carcinoma (SCC) often occurs in organs covered with squamous epithelium. Primary or metastatic SCC originating from the SI is very rare, with very few cases reported in the literature. CASE SUMMARY: This case report involves a 69-year-old man who developed abdominal pain after lunch. After admission, an abdominal computed tomography scan revealed perforation of the alimentary canal and multiple abnormal low-density lesions in the liver. During laparotomy, an approximately 4 cm × 3 cm-sized solid tumor was found in the jejunum, located 30 cm from the Treitz ligament, with a perforation. An intestinal segment of approximately 15 cm was removed, including the perforated portion. The pathological result was SCC. In combination with liver imaging, a diagnosis of SI SCC with multiple liver metastases was considered. The patient died from hepatic failure 1 mo after the operation. CONCLUSION: SI tumors are very rare compared to those originating in other digestive organs. Due to its insidious onset, the diagnosis of this disease is usually delayed. Clinicians must pay close attention to digestive symptoms such as persistent abdominal pain and melena.

17.
Aging (Albany NY) ; 14(9): 4137-4157, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35537818

ABSTRACT

BACKGROUND: We previously identified a novel lncRNA, CRART16, that could induce cetuximab resistance in colorectal cancer cells. This study explored the relationship of CRART16 expression to gastric cancer progression and the molecular mechanisms involved. METHODS: We evaluated CRART16 expression in gastric cancer tissues and adjacent normal tissues from the TCGA database and our hospital. Besides, we assessed its relationship with the overall survival (OS) of patients with gastric cancer. The effects of CRART16 on gastric cancer angiogenesis were determined by endothelial tube formation assay, spheroid sprouting assay, HUVEC invasion assay, and chick embryo chorioallantoic membrane (CAM) assay. The involvement of the lncRNA CRART16/miR-122-5p/FOS axis was analyzed by western blotting and dual-luciferase reporter assay. The functions of CRART16 were confirmed in xenograft mouse models. RESULTS: We found that CRART16 was substantially overexpressed in gastric cancer tissues compared with normal tissues, based on the TCGA database and our clinical samples. High expression of CRART16 correlated with more advanced tumor stages and poor prognosis. Overexpression of CRART16 in gastric cancer cells promoted proliferation, colony formation, angiogenesis, and bevacizumab resistance in vitro, and it promoted tumor growth and angiogenesis in vivo, and vice versa. CRART16 was found to downregulate miR-122-5p by acting as a sponge, upregulating the target oncogene FOS. Afterward, the increased FOS expression led to the upregulation of VEGFD. CONCLUSION: Our findings demonstrate that CRART16 promotes angiogenesis in vitro and in vivo, and CRART16 is a prognostic marker and therapeutic target in gastric cancer.


Subject(s)
MicroRNAs , Proto-Oncogene Proteins c-fos , RNA, Long Noncoding , Stomach Neoplasms , Vascular Endothelial Growth Factor D , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Chick Embryo , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor D/genetics
18.
J Int Med Res ; 50(3): 3000605221080679, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35277104

ABSTRACT

OBJECTIVE: The vascular anatomic variations of the right colon present a challenge for colorectal surgeons. However, there have been few detailed studies of the variations in the anterosuperior pancreaticoduodenal vein (ASPDV). METHODS: We studied consecutive patients with right colon cancer who underwent laparoscopic right hemicolectomy at Peking University First Hospital (N = 117) between January 2018 and June 2021. RESULTS: The variations in the ASPDV were classified as type I (n = 101, (86.3%)), defined as ASPDVs draining into the gastrocolic trunk of Henle (GCT); type II (n = 10, (8.5%)), defined as ASPDVs draining into the superior mesenteric vein (SMV); or type III, defined as ASPDVs draining into both the GCT and SMV. For type I, subtypes were defined according to the branching of the ASPDVs: subtype a, with one branch (n = 87, (86.1%)); subtype b, with two branches (n = 12, (11.9%)); and subtype c, with more than two branches (n = 2, (2.0%)). Type I was also subtyped according to the confluence of the ASPDV and GCT, with subtype 1 being defined by a proximal site (n = 96, 95%) and subtype 2 by a distal site (n = 5, 5.0%). CONCLUSIONS: We have characterized the variations in ASPDVs encountered during laparoscopic right hemicolectomy, which should provide a reference for colorectal surgeons.


Subject(s)
Laparoscopy , Mesenteric Veins , Colectomy , Colon/surgery , Humans , Mesenteric Veins/surgery , Retrospective Studies
19.
Oncogenesis ; 11(1): 12, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35256584

ABSTRACT

As one of the most common malignant tumors, it is particularly important to further understand the development mechanism of gastric cancer and to find more effective therapeutic target genes. The results of immunohistochemical staining showed that PSMC2 was upregulated in gastric cancer. Cell function experiments indicated that PSMC2 knockdown inhibited the proliferation, clone formation and migration of gastric cancer cells, and induced apoptosis. In vivo experiments further showed that PSMC2 knockdown suppressed tumor growth. RPS15A and mTOR pathway were identified the downstream gene and pathway of PSMC2 by GeneChip and IPA. PSMC2 knockdown inhibited RPS15A expression and mTOR pathway, which was neutralized by RPS15A overexpression. Overexpression of RPS15A promoted the proliferation and migration of gastric cancer cells, which alleviated the inhibitory effect caused by PSMC2 knockdown to a certain extent. The mTOR pathway inhibitor Torin1 partially restored the promoting role of RPS15A overexpression on the gastric cancer cell proliferation. Furthermore, bioinformatics analysis and dual-luciferase reporter assays showed that PSMC2 and RPS15A competitively bound to hsa-let-7c-3p. Inhibition of hsa-let-7c-3p promoted the migration of MGC-803 cells and reduced the apoptosis level, while simultaneous inhibition PSMC2 and hsa-let-7c-3p restored the migration and apoptosis levels of gastric cancer cells. In conclusion, PSMC2 and RPS15A were highly expressed in gastric cancer. PSMC2 enhanced RPS15A levels by targeting hsa-let-7c-3p, and then activated mTOR pathway, thereby promoting the progression of gastric cancer.

20.
Life Sci ; 292: 120330, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35051420

ABSTRACT

AIMS: This study investigated the protective effect of Escherichia coli Nissle 1917 (EcN) on intestinal barrier and the mechanism in the context of acute severe inflammation. MATERIALS AND METHODS: In this study, mice received lipopolysaccharide (LPS) intraperitoneal injection with or without EcN administration to construct a mouse model of endotoxemia. Clinical scores, intestinal permeability, inflammatory cytokines and histopathological analysis of four main organs from different groups were assessed. The expression of tight junction proteins and activation of RhoA/ROCK2/MLC signaling were examined using western blotting. The localization of tight junction proteins was examined by immunofluorescence. Caco-2 monolayers with or without TLR-4 knockdown were incubated with EcN or TNF-α/IFN-γ and the monolayer barrier function was assessed by transepithelial electrical resistance (TER) and FITC-dextran 4000 Da (FD-4) flux. The expression of tight junction proteins and activation of RhoA/ROCK2/MLC signaling were examined by western blotting. The localization of tight junction proteins was examined by immunofluorescence. KEY FINDINGS: We found that EcN downregulated the RhoA/ROCK2/MLC signaling pathway to preserve barrier function and alleviated systemic inflammation in mouse model. And EcN also protected barrier function of Caco-2 monolayers by inhibiting the activation of RhoA/ROCK2/MLC signaling via TLR-4. SIGNIFICANCE: The results indicated that EcN protected the intestinal barrier function in endotoxemia through inhibiting the activation of RhoA/ROCK2/MLC signaling via TLR-4.


Subject(s)
Endotoxemia/therapy , Escherichia coli/metabolism , Protective Agents/pharmacology , rhoA GTP-Binding Protein/metabolism , Animals , Caco-2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...