Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Front Plant Sci ; 13: 980840, 2022.
Article in English | MEDLINE | ID: mdl-36217333

ABSTRACT

Piedmont zones have been witnessing intensive human activities since ancient times. However, it remains unclear when it comes to the environmental mechanism for early humans exploiting piedmont zones. Here we present a case study about the interactions between early human activities and landscape evolution in the piedmont of Taihang Mountain, an area with prominent ecological and cultural significance in Chinese history. Based on chronological and pollen analyses, we reconstruct the regional landscape evolution in the Fengtougang (FTG) site of southern Taihang Mountain during the Holocene. The results show that the area has been dominated by terrestrial plants since the late Longshan culture (4000 BP), including a large number of Pinus, Artemisia, Spiraea, and Gramineae, a few Cattails, and some other aquatic herbs. During the early history (4000-2000 BP), there is a combination of Pinus, Artemisia, Spiraea, Compositae, and Selaginella Chinensis, with a few aquatic plants. Since the late history (500 BP), the Chinese selaginella, Pinus, Selaginella, and Sedge families dominate, with no aquatic plant pollen found. Combining the detailed geoarchaeological survey, grain size analysis, and magnetic susceptibility analysis, we demonstrate that there should be a landscape of extensive floodplain during the early-middle Holocene (10000-4000 BP). During the late Longshan culture (about 4000 BP), the study area should be dominated by a landscape of sparse forest grassland with interlacing rivers and lakes. With river downcutting and watercourse fixation since the late Longshan culture, the flooded area massively shrinks, providing suitable habitat for human settlement. From then on, human activities begin to move to the study area on a large scale, followed by continuous cultural development and thriving early civilization.

2.
J Cell Physiol ; 236(11): 7591-7604, 2021 11.
Article in English | MEDLINE | ID: mdl-34041752

ABSTRACT

Uncontrolled recurrence and metastasis are important reasons for the high mortality rate of malignant tumors. Vimentin is positively correlated with the degree of malignancy of cancer cells. Vimentin is also highly expressed in colorectal cancer (CRC) cells and plays a critical role in the metastasis and prognosis of CRC. However, the molecular mechanism of vimentin in the progression of CRC is incompletely understood. Therefore, the most active regions (nucleotides: 785-1085 nt) of the vimentin promoter in CRC were identified using luciferase experiments. By transcription factor sequence search and mutation analysis, the activator protein 1 (AP-1) binding site in the region of 785-1085 nt was confirmed. The vimentin promoter activity was enhanced by overexpression of AP-1. The electrophoretic mobility shift assay and chromatin immunoprecipitation assay showed that the binding site was recognized by AP-1. By cell proliferation assay, colony-forming assay, scratch-wound assay, cell migration assay, and cell invasion assay, we demonstrated that the AP-1 overexpression increased CRC cell proliferation, migration, and invasion. However, when vimentin was knocked down by vimentin small hairpin RNA in the CRC cell of AP-1 overexpression, this trend disappeared. Animal experiments and immunohistochemistry showed that AP-1 promoted tumor growth by regulating the vimentin gene. In summary, AP-1 affected metastasis, invasion of CRC cells in vitro, and tumor growth in vivo by activating the vimentin promoter. This study might provide new insights into the molecular mechanisms of the development of CRC and provide potential therapeutic targets for CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Transcription Factor AP-1/metabolism , Vimentin/metabolism , Animals , Binding Sites , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Promoter Regions, Genetic , Signal Transduction , Transcription Factor AP-1/genetics , Tumor Burden , Vimentin/genetics
3.
Sci Rep ; 11(1): 3316, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558632

ABSTRACT

Intricate ceramic bronze-casting moulds are among the most significant archaeological remains found at Bronze Age metallurgical workshops in China. Firing temperature was presumably one of the most important technical factors in mould making. However, it has proven difficult to determine the firing temperatures of excavated moulds using existing analytical methods. This study establishes an innovative new method for using Fourier-transform infrared spectroscopy (FTIR) to estimate the firing temperature of clay-containing remains. The method is based on the finding that the infrared absorptivity of fired clay minerals, measured at the Si-O-Si stretching resonance band, is negatively correlated with firing temperature. Moulds and mould cores dating to the Early Shang period (sixteenth to fourteenth century BCE) are found to have been fired at extremely low temperatures-as low as 200-300 °C in many instances. These results provide critical new data for understanding the metallurgical technology of ancient China.

4.
Onco Targets Ther ; 13: 11991-12001, 2020.
Article in English | MEDLINE | ID: mdl-33244241

ABSTRACT

BACKGROUND AND AIM: Some studies have confirmed that miRNA-140 exhibits a suppressive role in gastric cancer, Wilms' tumor. However, the function of miRNA-140 in colorectal cancer has not been completely elucidated. The present study aims to verify TRAF6 as the targeted gene by miRNA-140 which was investigated in colorectal cancer tissues and cells, and its effects on the biological characteristics of colorectal cancer cells were determined, in order to provide an experimental and theoretical basis for the application of TRAF6 in the treatment of colorectal cancer. METHODS: qPCR analyzed miRNA-140 expression levels in colorectal cancer tissues, normal colorectal cancer tissues and colorectal cells including SW480 and HCT116 cancer cells and FHC normal colorectal epithetical cells. A serial biological experiment analyzed miRNA-140 effects on cell proliferation, migration and invasion capacities in SW480 and HCT116 cells. miRNA targeting gene prediction and a dual luciferase assay were used to analyze miRNA-140-targeted TRAF6. qPCR and Western blot analyzed miRNA-140 effects on the mRNA and protein expression of TRAF6. Western blot analyzed miRNA-140 effects on NF-κB/c-jun signaling pathways. Animal studies were performed to investigate the effects of miRNA-140 on colorectal cancer implantation tumor growth. Immunohistochemistry analyzed TRAF6 expression in animal experimentation tumors. RESULTS: miRNA-140 expression is lower in colorectal cancer tissues and colorectal cancer cells. Over-expression of miRNA-140 inhibited the proliferation, migration and invasion capacities of colorectal cancer cells. miRNA-140 targeted the TRAF6 mRNA 3'UTR area and decreased TRAF6 protein expression. miRNA-140 suppressed p-NF-κB/p-c-jun proteins expression. miRNA-140 inhibited colorectal cancer implantation tumor growth in the mice model. CONCLUSION: miRNA-140 targeting TRAF6 affects the progression and growth of colorectal cancer, the mechanism could be miRNA-140 decreasing the TRAF6 expression effects on the NF-κB/c-jun signaling pathways.

5.
Saudi J Gastroenterol ; 26(1): 46-52, 2020.
Article in English | MEDLINE | ID: mdl-32031158

ABSTRACT

BACKGROUND/AIMS: Gastric signet ring cell carcinoma (GSRC), a subtype of adenocarcinoma, has been considered a histological type with poor survival. We aimed to compare the survival outcomes between patients with GSRC and patients with gastric non-signet ring cell adenocarcinoma (NGSRC) and constructed a nomogram to predict gastric adenocarcinoma-specific survival (GCSS). PATIENTS AND METHODS: We identified 10,031 patients with gastric adenocarcinoma (GA) from the surveillance, epidemiology, and end results (SEER) database and stratified them into two histological type groups: GSRC and NGSRC. We used propensity score matching and identified 4304 patients (training cohort) to assess the effect of the histological type on GCSS with Kaplan-Meier curves, and constructed a predictive nomogram. The accuracy of the nomogram was tested on the remaining 5727 patients (validation cohort) with concordance index (C-index) values, calibration curves, and receiver operating characteristic (ROC) curve analysis. RESULTS: We found that the histological type SRC was not associated with significantly poor survival (5-year survival rate: 46.1% vs 46.7%, P = 0.822). GSRC patients had similar GCSS rates compared to those with NGSRC in each tumor, node, and metastasis (TNM) stage (allP > 0.05). The nomogram showed that histological type was a relatively weak predictor of survival. The C-index value of the nomogram for predicting survival was 0.720, similar to that in the validation cohort (0.724). CONCLUSIONS: Patients with GSRC had a similar prognosis to those with NGSRC. The proposed nomogram allowed a relatively accurate survival prediction for operable GA patients after gastrectomy.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Signet Ring Cell/pathology , Histology/classification , Stomach Neoplasms/pathology , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Aged , Carcinoma, Signet Ring Cell/mortality , Carcinoma, Signet Ring Cell/surgery , Case-Control Studies , Female , Gastrectomy/methods , Gastrectomy/mortality , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasm Staging , Nomograms , Predictive Value of Tests , Prognosis , Risk Factors , Stomach Neoplasms/epidemiology , Survival Rate , United States/epidemiology
6.
Transl Cancer Res ; 9(2): 910-917, 2020 Feb.
Article in English | MEDLINE | ID: mdl-35117436

ABSTRACT

BACKGROUND: Many studies have been performed to evaluate the effect of subcutaneous suction drainage to prevent incisional surgical site infections (SSIs) after radical colorectal surgery. However, the result has been controversial. The main reason may be that subcutaneous suction drainage is more prone to develop blockages, and the drainage tubes themselves serve as a conduit for bacteria into the wound. Therefore, we modified this method and evaluated this new method (subcutaneous suction drainage and intermittent irrigation) in patients who underwent radical colorectal surgery. METHODS: A total of 119 patients who underwent open radical colorectal surgery were included in our study from April 2015 to November 2017. A total of 61 patients were included in the irrigation group (subcutaneous suction drainage or intermittent irrigation), and 58 patients were included in the control group (no subcutaneous suction drainage and intermittent irrigation). The key endpoints were the incidence rate of incisional SSIs, the inpatient stay, and hospitalization expenses. All of the patients in our study had the following characteristics: (I) their subcutaneous fat thickness was more than 1.5 cm by means of CT or MRI measure before operation; (II) the patients had at least one of the following cases before operation: diabetes mellitus, hypoalbuminemia (ALB ≤35 g/L), anemia (Hb ≤90 g/L) or tumorous obstruction. RESULTS: The incidence of incisional SSIs rate was 27/119 (22.7%) in the overall patients, 22/61 (36.1%) in the control group, and 5/58 (8.6%) in the group. The rate of SSIs in the irrigation group was significantly lower than the control group (P<0.001). The inpatient stay (9.64±4.15) in the irrigation group was shorter than the control group (12.26±5.55) (P=0.004). The hospitalization expenses (57,356±9,518) in the irrigation group were lower than the control group (62,119±11,101) (P=0.014). One of the patients in the control group died of pulmonary infection due to intraoperative aspiration. There was no death in the irrigation group. CONCLUSIONS: The subcutaneous suction drainage and intermittent irrigation is safe and effective to prevent incisional SSIs in radical colorectal surgery.

7.
Cancer Genomics Proteomics ; 17(1): 23-33, 2020.
Article in English | MEDLINE | ID: mdl-31882548

ABSTRACT

BACKGROUND/AIM: Colorectal cancer (CRC) cells secrete inflammatory cytokines that affect CRC progression. The aim of the present study was to determine if micro-RNA-140(miR-140) regulates inflammatory cytokine secretion induced by lipopolysaccharide (LPS) in colorectal cancer cells by targeting tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6). MATERIALS AND METHODS: Fifty fresh colon-cancer specimens and normal colorectal tissues were collected from patients with CRC and tested for the expression miR-140. Human CRC cell lines SW480 and HCT116 were treated with various concentrations and times with LPS. miR-140 and mRNA expression of potentially related genes were analyzed by qPCR. Protein expression was analyzed using western blot or ELISA. Overexpression plasmids with pcDNA3.1-TRAF6, pGL4.10-wtTRAF6 and pGL4.10-mutTRAF6 were constructed. miRNA target gene prediction and a dual luciferase assay were used to analyze miR-140-targeted TRAF6. RESULTS: miR-140 expression was up-regulated in CRC tissues. In CRC cells, LPS could increase miR-140 expression in a time- and concentration-dependent manner. LPS increased inflammatory cytokine mRNA expression levels in SW480 and HCT116 human colon-cancer cells. miRNA-140 suppressed TRAF6 expression via targeting the 3'UTR. TRAF6 affected miR-140-mediated inflammatory cytokine expression of SW480 and HCT116 cells under LPS treatment. CONCLUSION: miR-140 regulates inflammatory cytokine secretion of LPS-induced colorectal cancer cells by targeting TRAF6.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Cytokines/metabolism , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Prognosis , Signal Transduction , Tumor Cells, Cultured
8.
Biol Chem ; 401(2): 285-296, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31188741

ABSTRACT

The gene encoding transmembrane protein 100 (TMEM100) was first discovered to be transcribed by the murine genome. It has been recently proven that TMEM100 contributes to hepatocellular carcinoma and non-small-cell lung carcinoma (NSCLC). This study investigates the impact of TMEM100 expression on gastric cancer (GC). TMEM100 expression was remarkably downregulated in GC samples compared to the surrounding non-malignant tissues (p < 0.01). Excessive TMEM100 expression prohibited the migration and invasion of GC cells without influencing their growth. However, TMEM100 knockdown restored their migration and invasion potential. Additionally, TMEM100 expression restored the sensitivity of GC cells to chemotherapeutic drugs such as 5-fluouracil (5-FU) and cisplatin. In terms of TMEM100 modulation, it was revealed that BMP9 rather than BMP10, is the upstream modulator of TM3M100. HIF1α downregulation modulated the impact of TMEM100 on cell migration, chemotherapy sensitivity and invasion in GC cells. Eventually, the in vivo examination of TMEM100 activity revealed that its upregulation prohibits the pulmonary metastasis of GC cells and increases the sensitivity of xenograft tumors to 5-FU treatment. In conclusion, TMEM100 serves as a tumor suppressor in GC and could be used as a promising target for the treatment of GC and as a predictor of GC clinical outcome.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Membrane Proteins/genetics , Stomach Neoplasms/drug therapy , Animals , Biomarkers, Tumor/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Screening Assays, Antitumor , Female , Fluorouracil/pharmacology , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured
9.
Int J Mol Med ; 45(1): 131-140, 2020 01.
Article in English | MEDLINE | ID: mdl-31746347

ABSTRACT

The role of myeloid differentiation factor 88 (MyD88) in malignant tumors is largely unknown. Therefore, in this study, we aimed to examine the function and underlying mechanism of MyD88 in colorectal carcinoma in vitro using SW480 and HCT116 cell lines and in vivo using a nude mouse model. SW480 and HCT116 cells were infected with a lentiviral­based effective MyD88 siRNA virus. CCK­8 and colony formation assay were used to assess cell proliferation. Transwell and scratch assays were used to test the migration of colorectal cancer cells, and the Transwell assay was further used to analyze the invasiveness of colorectal cancer cells. Western blotting was performed to analyze the underlying mechanism of MyD88 regulation. In vitro experiments demonstrated that silencing MyD88 in SW480 and HCT116 cells markedly suppressed growth and invasion. Furthermore, MyD88 knockdown affected the MyD88­NF­κB/AP­1 signaling pathways in SW480 and HCT116 cells. In vivo, MyD88 knockdown inhibited tumor growth in a HCT116 cell subcutaneous nude model. We found that knockdown of the MyD88 gene can affect proliferation, invasion, and migration of colorectal cancer cells. We further verified that MyD88 knockdown can reduce the activity of NF­κB and AP­1 pathways. These results show that MyD88 gene plays an important role in promoting colorectal cancer, and thus can be exploited as a potential diagnostic and prognostic biomarker for colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Knockdown Techniques , Heterografts , Humans , Mice , Myeloid Differentiation Factor 88/metabolism , RNA Interference , RNA, Small Interfering/genetics
10.
Cancer Genomics Proteomics ; 16(6): 409-419, 2019.
Article in English | MEDLINE | ID: mdl-31659096

ABSTRACT

BACKGROUND/AIM: Inflammation may play a role in cancer initiation and progression. The molecular mechanisms by which inflammation causes colorectal cancer, remains unclear. The present study investigated a signaling pathway that affects inflammation in colorectal cancer. MATERIALS AND METHODS: SW480 cells, HCT116 cells, and cells with knockdown of myeloid differentiation 88 (MyD88), and forced expression of MyD88 were treated with lipopolysaccharide (LPS; 1 µg/ml). Inflammation-related mRNA expression was analyzed by the quantitative reverse transcription polymerase chain reaction and inflammatory cytokines were detected by western blotting. The enzyme-linked immunosorbent assay (ELISA) was used to quantify inflammation-related cytokines in colorectal cancer cells. Cancer cell properties were evaluated using the wound-healing assay, transwell migration assay, transwell invasion assay, colony-formation assay, and CCK-8 assay. RESULTS: LPS up-regulated mRNA and protein levels of inflammatory factors in colorectal cancer cells. Knockdown of MyD88 inhibited LPS-induced mRNA expression and inflammatory protein expression in colorectal cancer cells. Similarly, silencing of MyD88 expression suppressed LPS-induced changes in the biological behavior of colorectal cancer cells. Silencing of MyD88 expression down-regulated expression of proteins of the LPS/nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-ĸB)/mitogen-activated protein kinase (MAPK) signaling pathway. Restoration of the expression of MyD88 reversed the effects in LPS-treated HCT116 cells. CONCLUSION: MyD88-regulated LPS/NF-ĸB/MAPK signaling pathway affects the inflammatory and biological behavior of LPS-induced colorectal cancer cells.


Subject(s)
Colorectal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Myeloid Differentiation Factor 88/biosynthesis , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , HCT116 Cells , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Neoplasm Proteins/genetics
11.
Life Sci ; 235: 116831, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31487530

ABSTRACT

AIMS: TRAF6 is an intracellular signal adapter molecule plays a significant role in tumor development. However, the specific mechanism causes and promotes of colorectal cancer keep largely unknown. Therefore, we sought to investigate the roles and the molecular mechanisms of TRAF6 in regulation colorectal cancer. MATERIAL AND METHODS: The immunohistochemistry analyzed the expression of TRAF6 in colorectal cancer samples and analyzed the effects of expression of TRAF6 on the prognosis in colorectal cancer. The roles of TRAF6 in regulating colorectal cancer cell proliferation, colony formation, cell migration, cell wound healing and cell invasion were evaluated in vitro. Animal studies were performed to investigate the effects of TRAF6 on tumor growth. mRNA abundance of key genes was analyzed via qPCR. Protein level of TRAF6 and NF-κB/AP-1 signaling pathways was examined by Western blot. Luciferase reporter and Immunofluorescence assays were used to identify the activities NF-κB/AP-1 signaling pathways. KEY FINDINGS: TRAF6 high expression in colorectal cancer tissues. And colorectal cancer patients with high expression of TRAF6 had a poor survival rate. TRAF6 knockdown can inhibit proliferation, migration, and invasion of colorectal cancer cells in vitro and in vivo experiments. TRAF6 activates the TRAF6-NF-κB/AP-1 signaling pathway by entering the nucleus, causing biobehavioral changes in colorectal cancer cells. SIGNIFICANCE: TRAF6 plays a vital role in the progression of colorectal cancer. What's more, research elucidating the biological mechanisms of TRAF6 can treated as potential therapeutic target for colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Adult , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colorectal Neoplasms/physiopathology , Disease Progression , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/physiopathology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/antagonists & inhibitors , TNF Receptor-Associated Factor 6/physiology , Tumor Stem Cell Assay , Wound Healing/physiology
12.
World J Clin Cases ; 6(15): 985-994, 2018 Dec 06.
Article in English | MEDLINE | ID: mdl-30568953

ABSTRACT

AIM: To perform a meta-analysis to investigate the correlation between body mass index (BMI) and the short-term outcomes of laparoscopic gastrectomy (LG) for gastric cancer (GC) in Asian patients. METHODS: The PubMed, Cochrane, EMBASE, and Web of Science databases were searched for studies that focused on the impact of obesity on the short-term outcomes of LG for GC in Asian patients who were classified into a high BMI (BMI ≥ 25 kg/m2) or low BMI group (BMI < 25 kg/m2). The results are expressed using the pooled odds ratio (OR) for binary variables and standard mean difference (SMD) for continuous variables with 95% confidence interval (CI), and were calculated according to the fixed-effects model while heterogeneity was not apparent or a random-effects model while heterogeneity was apparent. RESULTS: Nine studies, with a total sample size of 6077, were included in this meta-analysis. Compared with the low BMI group, the high BMI group had longer operative time (SMD = 0.26, 95%CI: 0.21 to 0.32, P < 0.001), greater blood loss (SMD = 0.19, 95%CI: 0.12 to 0.25, P < 0.001), and fewer retrieved lymph nodes (SMD = -0.13, 95%CI: 0.18 to 0.07, P < 0.001). There was no significant difference between the high and low BMI groups in postoperative complications (OR = 1.12, 95%CI: 0.95 to 1.33, P = 0.169), the duration of postoperative hospital stay (SMD = 0.681, 95%CI: -0.05 to 0.07, P = 0.681), postoperative mortality (OR = 1.95, 95%CI: 0.78 to 4.89, P = 0.153), or time to resuming food intake (SMD = 0.00, 95%CI: -0.06 to 0.06, P = 0.973). CONCLUSION: Our meta-analysis provides strong evidence that despite being associated with longer operative time, greater blood loss, and fewer retrieved lymph nodes, BMI has no significant impact on the short-term outcomes of LG for GC in Asian patients, including postoperative complications, the duration of postoperative hospital stay, postoperative mortality, and time to resuming food intake. BMI may be a poor risk factor for short-term outcomes of LG. Other indices should be taken into account.

13.
Front Biosci (Landmark Ed) ; 22(2): 248-257, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814614

ABSTRACT

Accumulating evidence suggests that the anti-diabetic drug, metformin, exerts anti-proliferative effects in many types of cancers. However, the function and mechanisms of metformin in human colorectal cancer (CRC) remain unknown. Here, we show that metformin induces growth inhibition and apoptosis through activating AMPK-mTOR pathway in human colorectal cancer cells. Notably, metformin treatment significantly up-regulated adenosine A1 receptor (ADORA1) expression in human colorectal cancer cells, while suppression of ADORA1 activity by its specific inhibitor rescued the growth inhibition induced by metformin. Moreover, ADORA1-mediated growth inhibition and apoptosis induced by metformin is AMPK-mTOR pathway dependent in human colorectal cancer cells. Taken together, these results indicate that metformin suppresses human colorectal cancer growth by inducing apoptosis via ADORA1, which provide evidence the anti-neoplastic effects of metformin in the treatment of human colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Metformin/pharmacology , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Receptor, Adenosine A1/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects
14.
Oncotarget ; 7(45): 73711-73724, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27713159

ABSTRACT

Lipopolysaccharide (LPS) exists in the outer membrane of Gram-negative bacteria. Colorectal normal epithelium and colorectal cancer cells in situ are continuously exposed to LPS from intestinal bacteria, while little is known about the influence of LPS on colorectal cancer progression and metastasis. In this study, we investigated the potential role of LPS on colorectal cancer progression and metastasis as well as the underlying mechanisms. We measured higher LPS concentration in colorectal cancer tissues and even higher LPS concentration in colorectal cancer tissues with lymph node metastasis. LPS significantly enhanced cancer cell motility and promoted human dermal lymphatic endothelial cells' (HDLECs') capacity of tube-like formation in vitro, as well as accelerates lymphangiogenesis and lymph node metastasis in nude mice. Furthermore, we demonstrated LPS notably increased the expression of VEGF-C in a time-dependent and concentration-dependent manner. VEGF-C is a key regulator for lymphangiogenesis and lymph node metastasis. By constructing lentivirus-mediated shVEGF-C cells, VEGF-C down-regulation suppressed LPS' promotive effect on cancer cell motility and HDLEC tube-like formation capacity. In addition, we found TLR4- NF-κB/JNK signal pathways were important for LPS to increase VEGF-C expression. All these result suggested a critical role for LPS in migration, invasion, lymphangiogenesis and lymph node metastasis of colorectal cancer, providing evidence that LPS increased VEGF-C secretion to promote cell motility and lymphangiogenesis via TLR4- NF-κB/JNK signaling.


Subject(s)
Colorectal Neoplasms/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Lymphangiogenesis/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Vascular Endothelial Growth Factor C/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphatic Metastasis , Promoter Regions, Genetic , Vascular Endothelial Growth Factor C/genetics
15.
Cell Physiol Biochem ; 39(5): 1665-1678, 2016.
Article in English | MEDLINE | ID: mdl-27639612

ABSTRACT

BACKGROUND AND AIM: Lipopolysaccharide(LPS) could promote the progression of colorectal cancer, but the specific regulatory mechanisms are largely unknown. So, this study aim to clarify the mechanisms that LPS upregulated VEGFR-3, which promotes colorectal cancer cells migration and invasion with a mechanism of increased NF-κB bind to the promoter of VEGFR-3. METHODS: The present study examined the VEGFR-3 expression in colorectal cancer tissues and analyzed the relationship between the VEGFR-3 expression with clinical parameters. PCR, Western blot, CCK-8, colone formation assay, and Transwell assay detected that LPS promoted the migration and invasion and the role of VEGFR-3 in the process of colorectal carcinoma in vitro. Used the methods of promoter analysis, EMSA assay and ChIP assay to explore the mechanisms LPS increased the expression of VEGFR-3. RESULTS: VEGFR-3 was significantly high expression in the colorectal cancer tissues. And the high expression was associated with the TNM stage and lymph node metastasis of colorectal cancer. LPS could promote the migration and invasion, which could be blocked by the neutralizing antibody IgG of VEGFR-3. And found that -159 nt to +65 nt was the crucial region of VEGFR-3 promoter. And detected that the NF-κB was important transcription factor for the VEGFR-3 promoter. And LPS could increase NF-κB binding to VEGFR-3 promoter and upregulated the expression of VEGFR-3 to exert biological functions. CONCLUSION: We have elucidated the relationship between LPS and the VEGFR-3 expression and revealed that VEGFR-3 play very important role in the process of LPS promoting the migration and invasion of colorectal cancer cells. Further illuminated the mechanism that LPS upregulated VEGFR-3 expression via increased NF-κB bind to the promoter of VEGFR-3.


Subject(s)
Cell Movement/drug effects , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lipopolysaccharides/pharmacology , NF-kappa B/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Aged , Antibodies, Neutralizing/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , HCT116 Cells , Humans , Immunoglobulin G/pharmacology , Male , Middle Aged , NF-kappa B/immunology , Neoplasm Invasiveness , Neoplasm Staging , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/immunology
16.
Mol Clin Oncol ; 5(2): 257-262, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446560

ABSTRACT

A discrepancy exists between the 7th edition guidelines of the American Joint Committee on Cancer (AJCC) and the 3rd edition Japanese treatment guidelines in terms of the classification of No. 12a lymph nodes as regional or distant lymph nodes in D2 lymphadenectomy for gastric cancer. The scope definition of No. 12a lymph nodes has yet to be fully elucidated. The present study aimed to assess the appropriateness of reclassifying No. 12a lymph node metastasis as distant metastasis according to the survival rate outcome, and to provide a clear and practical definition of the No. 12a group lymph nodes of gastric cancer. A retrospective analysis was performed on patients with gastric cancer who underwent standard or greater lymphadenectomy between January 2000 and December 2009 to find an association between No. 12a node metastasis and survival outcome. The present study first presented a clear and practical scope definition of the No. 12a group lymph nodes of gastric cancer, according to our clinical experiences and practices (Table I and Fig. 1). The survival outcome of patients with gastric cancer and No. 12a lymph node metastasis was poorer compared with that of patients with no No. 12a lymph node metastasis (P=0.0003). The results were similar in stage III patients with gastric cancer (P<0.0001). However, the survival outcome of patients was similar with or without No. 12a lymph node metastasis in stage IV gastric cancer (P=0.1968). Cox regression analysis revealed that the AJCC stage was independently associated with an unfavorable cumulative survival rate. Logistic regression analysis revealed that tumor location, AJCC stage, intravascular cancer emboli and nerve invasion were associated with No. 12a lymph node metastasis. In conclusion, the data in the present study suggested that No. 12a lymph node metastasis is associated with distant metastasis, and therefore they concur with the 7th edition AJCC gastric cancer guidelines, which appear to be correct in terms of considering No. 12a lymph node metastasis as distant metastasis.

17.
Int J Oncol ; 48(6): 2387-98, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27035122

ABSTRACT

The receptor-interacting protein-1 (RIP-1) is an important molecular in inflammation signaling pathways, but the role of RIP-1 in gastric cancer is largely unknown. In this study, we tested the expression of RIP-1 in gastric cancer samples and analyzed the effects of expression of RIP-1 on the prognosis in gastric cancer patients. We analyzed the role of the RIP-1 in gastric cancer cells and addressed the functional role of RIP-1 using a xenograft mouse model. A lentivirus-based effective RIP-1 siRNA vector was infected into HGC and AGS cells. The effect of RIP-1 siRNA on HGC and AGS cells were investigated by cell proliferation assay and invasion assay. Furthermore, we examined the role of RIP-1-siRNA on HGC cells in the mice with subcutaneous xenograft tumor, and preliminarily analyzed the underlying mechanisms. The results indicated that the expression of RIP-1 in the gastric cancer tissues was significantly higher than the expression in the normal gastric tissues. Additionally, RIP-1 immunoreactivity was positive at the site of invasion, but little or no immunoreactivity was detected at the gastric cancer parts of interstitial substance. Gastric cancer patients with high expression of RIP-1 had a poor survival rate. RIP-1 expression in the gastric cancer cell lines were general. HGC-R-1-RNAi-LV inhibited HGC and AGS cell proliferation and invasion ability in vitro. RIP-NF-κB/AP-1-VEGF-C signaling pathways have a crucial role in the regulate the biological functions of HGC cells. HGC-R-1-RNAi-LV suppressed tumor growth in the HGC cell subcutaneous xenograft model. In conclusion, our data indicate that RIP-1 promote the growth and invasion of gastric cancer in vitro and in vivo, additionally providing evidence that targeting RIP-1 may be useful in the treatment of gastric cancer.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Case-Control Studies , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Nuclear Pore Complex Proteins/genetics , RNA, Small Interfering , RNA-Binding Proteins/genetics , Stomach Neoplasms/genetics , Transfection
18.
Oncol Rep ; 35(3): 1787-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26750536

ABSTRACT

Gastric cancer shows the highest invasive and metastasis features, especially lymph metastasis, which is closely associated with poor prognosis of gastric cancer. Although there is evidence that interleukin-6 (IL-6) can promote gastric cancer progression, the underlying specific mechanisms and the mechanisms of gastric cancer lymphangiogenesis are largely unknown. In the present study, we explore whether IL-6 could promote the proliferation and invasion activity of gastric cancer cells, and whether IL-6 mediating VEGF-C production affected the lymphangiogenesis in gastric cancer cells. Our results revealed that IL-6 and its receptors (IL-6 and gp130) are broadly expressed in various gastric cancer cell lines including SGC-7901, MGC, MKN-28 and AGS. Exogenous IL-6 increased the ability of gastric cancer cell proliferation and invasion, which could be weakened by AG490. in addition, exogenous IL-6 promoted the VEGF-C production of gastric cancer cells and the lymphangiogenesis of HDLECs. As we expected, AG490 was able to reduce these effects. Western blot analysis showed that IL-6 increased JKA, STAT3, p-STAT3 and VEGF-C protein levels in the gastric cancer cells. However, the JKA, STAT3, p-STAT3 and VEGF-C protein expression levels were inhibited by AG490. Our data suggested that IL-6 mediates the singnal pathway of JAK-STAT3-VEGF-C promoting the growth, invasion and lymphangiogenesis in gastric cancer. Thus, IL-6 and its related signal pathways may be a promising target for treatment of gastric cancer growth and lymphangiogenesis.


Subject(s)
Interleukin-6/genetics , Lymphangiogenesis/genetics , STAT3 Transcription Factor/genetics , Stomach Neoplasms/genetics , Vascular Endothelial Growth Factor C/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cytokine Receptor gp130/biosynthesis , Cytokine Receptor gp130/genetics , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/biosynthesis , Janus Kinases/genetics , Lymphatic Metastasis , Neoplasm Invasiveness/genetics , STAT3 Transcription Factor/biosynthesis , Signal Transduction , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor C/biosynthesis
19.
Tumour Biol ; 35(2): 1331-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24234331

ABSTRACT

Previous studies have assessed the prognostic role of serum carbohydrate antigen 19-9 (CA 19-9) concentration in patients with gastric cancer, but the findings from those studies were inconsistent. We searched the PubMed and Web of Science databases to find eligible studies assessing the prognostic role of CA 19-9 in patients with gastric cancer. Twelve studies with a total of 5,072 gastric cancer patients were finally included into the meta-analysis. The pooled hazard ratio (HR) with corresponding 95 % confidence interval (95 % CI) for overall survival were calculated to assess the prognostic role of CA 19-9 in patients with gastric cancer. Overall, elevated serum concentration of CA 19-9 (>37 U/mL) was associated with poorer overall survival in patients with gastric cancer (fixed-effects HR = 1.36, 95 % CI 1.24-1.48, P < 0.001). Subgroup analysis by study design further showed that elevated serum concentration of CA 19-9 was associated with poorer overall survival in patients with gastric cancer. There was no obvious risk of publication bias. Elevated concentration of serum CA 19-9 is associated with poorer overall survival in patients with gastric cancer.


Subject(s)
CA-19-9 Antigen/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Stomach Neoplasms/genetics , CA-19-9 Antigen/blood , Humans , Prognosis , Risk Factors , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...