Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 127(5): 811-823, 2022 09.
Article in English | MEDLINE | ID: mdl-35637411

ABSTRACT

BACKGROUND: Multiple mechanisms have been proposed that lead to reduced effectiveness of trastuzumab in HER2-positive gastric cancer (GC), yet resistance to trastuzumab remains a challenge in clinics. METHODS: We established trastuzumab-resistant cells and patient-derived xenografts models to measure metabolic levels and vascular density and shape. The HER2-positive GC patient samples were used to determine clinical significance. We also measured protein expression and phosphorylation modifications to determine those alterations related to resistance. In vivo studies combining inhibitor of PFKFB3 with trastuzumab corroborated the in vitro findings. RESULTS: The 6-phosphofructo-2-kinase (PFKFB3)-mediated trastuzumab resistance pathways in HER2-positive GC by activating the glycolytic pathway. We also found vessels are chaotic and destabilised in the tumour during the trastuzumab resistance process. Inhibition of PFKFB3 significantly diminished tumour proliferation and promoted vessel normalisation in the patient-derived xenograft model. Mechanistically, PFKFB3 promoted the secretion of CXCL8 into the tumour microenvironment, and phosphorylated Ser1151 of ERBB2, enhancing the transcription of CXCL8 by activating the PI3K/AKT/NFκB p65 pathway. CONCLUSIONS: Our current findings discover that PFKFB3 inhibitors might be effective tools to overcome adjuvant therapy resistance in HER2-positive GC and reshaping the microenvironment by normalising tumour vessels is a novel strategy to overcome trastuzumab resistance.


Subject(s)
Phosphofructokinase-2 , Stomach Neoplasms , Trastuzumab , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/genetics , Phosphofructokinase-2/pharmacology , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Tumor Microenvironment
2.
Arch Biochem Biophys ; 722: 109216, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35421356

ABSTRACT

BACKGROUND: FAM98A is a microtubule-associated protein involved in cell proliferation and migration, and is frequently dysregulated in epithelial cancers. But its role in the development of colorectal cancer (CRC) cancer remains unknown. METHODS: Immunohistochemical analysis was performed to examine the expression of FAM98A in CRC samples. We also investigated the effects of abnormal expression on the biological behavior of colorectal cancer cells both in vitro and in vivo. Immunoblotting and immunoprecipitation were used to screen FAM98A-related signalling pathways and downstream factors. RESULTS: FAM98A was upregulated in CRC tissues and CRC cell lines. Overexpression of FAM98A promoted cell proliferation and recovered 5-FU suppressed CRC cell proliferation both in vitro and in vivo. In addition, the Enhanced expression of FAM98A inhibited ferroptosis in CRC cells by activating the translation of xCT in stress granules (SGs). Furthermore, we identified that metformin could reverse FAM98A-mediated 5-FU resistance in CRC cells. CONCLUSIONS: Our results for the first time indicate that FAM98A plays a critical role in promoting CRC progression, which provides a novel target for clinical drug resistance of colorectal cancer. And metformin may sensitize 5-FU in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Metformin , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Proteins/metabolism
3.
Arch Biochem Biophys ; 702: 108838, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33727040

ABSTRACT

The antimetabolite 5-fluorouracil (5-FU) is a widely used chemotherapy regimen for the treatment of gastric cancer (GC). However, resistance to 5-FU remains a major drawback in the clinical use. The treatments of anti-tumor chemo-agents recruit tumor associated macrophages (TAMs) which are highly implicated in the chemoresistance development, but the underlying molecular mechanism is unclear. Here, we demonstrate that YAP1 is overexpressed in resistant GC tissues compared to sensitive GC tissues. Further, IL-3 secreted by YAP1-overexpressed GC could skew macrophage polarization to M2-like phenotype and inducing GLUT3-depended glycolysis program. Meanwhile, polarized M2 macrophages enhance 5-FU resistance in tumor cells by secreting CCL8 and activating phosphorylation of JAK1/STAT3 signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Glucose Transporter Type 3/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Carcinogenesis/drug effects , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Humans , Male , Middle Aged , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , YAP-Signaling Proteins
4.
Cancer Cell Int ; 20: 503, 2020.
Article in English | MEDLINE | ID: mdl-33061855

ABSTRACT

BACKGROUND: Tumors display a high rate of glucose metabolism and the SLC2A (also known as GLUT) gene family may be central regulators of cellular glucose uptake. However, roles of SLC2A family in mechanism of metabolite communication with immunity in gastric cancer remains unknown. METHODS: Bioinformatics analysis and IHC staining were used to reveal the expression of SLC2A3 in gastric cancer and the correlation with survival prognosis. Real-time PCR, western blots, OCR, ECAR, lactate production and glucose uptake assays were applied to determine the effect of SLC2A3 on glycolysis reprogramming. We then investigated the consequences of SLC2A3 upregulation or inhibition on aerobic glycolysis, also explored the underlying mechanism. Bioinformatics analysis and in vitro and in vivo research were used to reveal the role of SLC2A3 in macrophage infiltration and transition. RESULTS: Here, we show that SLC2A3 acts as a tumor promoter and accelerates aerobic glycolysis in GC cells. Mechanistically, the SLC2A3-STAT3-SLC2A3 feedback loop could promote phosphorylation of the STAT3 signaling pathway and downstream glycolytic targeting genes. Moreover, SLC2A3 potentially contributes to M2 subtype transition of macrophage infiltration in the GC microenvironment. CONCLUSIONS: SLC2A3 could be used as a prognostic biomarker to determine prognosis and immune infiltration in GC and may provide an intervention strategy for GC therapy.

5.
Chin Med J (Engl) ; 133(15): 1805-1814, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32649507

ABSTRACT

BACKGROUND: We previously found that the intestinal epithelial chemokine (C-C motif) ligand 7 (CCL7) plays an important role in the development of toxin-induced acute liver damage. The detailed effects of intestinal epithelial CCL7 on chronic diseases; however, are still unclear. Here, we aimed to investigate the impact of intestinal epithelial CCL7 overexpression on high-fat diet (HFD)-induced obesity and steatohepatitis in mice. METHODS: Intestinal epithelial CCL7 overexpression (CCL7) mice and their wild-type (WT) littermates were fed with normal chow or HFD for 16 weeks to induce obesity and non-alcoholic fatty liver disease. Body weight gain, as well as adipose tissue index were assessed. Liver injury was monitored by histological analysis and real time polymerase chain reaction. Gut microbial composition was analyzed by 16S rRNA gene sequencing. RESULTS: We found that the CCL7 mice on a HFD had markedly decreased weight gain (8.9 vs. 17.0 g, P < 0.05) and a lower adipose tissue index that include mesenteric fat (1.0% vs. 1.76%, P < 0.05), gonadal fat (2.1% vs. 6.1%, P < 0.05), subcutaneous fat (1.0% vs. 2.8%, P < 0.05) compared to WT animals. HFD-induced glucose intolerance and insulin resistance were also significantly improved in CCL7 mice compared to WT. Furthermore, HFD-fed CCL7 mice displayed less hepatic lipid accumulation and lower expression of inflammatory factors than WT mice. 16S rRNA gene sequencing demonstrated that CCL7 overexpression in intestinal epithelial cells improved HFD-induced gut microbial dysbiosis. CONCLUSIONS: Our study revealed that CCL7 overexpression in the intestinal epithelium protects mice against the progression of diet-induced obesity, hepatic steatosis, and enteric dysbiosis.


Subject(s)
Gastrointestinal Microbiome , Insulin Resistance , Animals , Chemokines , Diet, High-Fat/adverse effects , Ligands , Liver , Mice , Mice, Inbred C57BL , Obesity/genetics , RNA, Ribosomal, 16S
6.
J Transl Med ; 18(1): 276, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32641074

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

7.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1777-1778, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32444990

ABSTRACT

After re-read our published article, the authors found a mistake and would like to make correction: Fig. 4a, b, 0h groups, we mistakenly placed wrong representative staining pictures in the original figure, the correct figures are showed as follow.

8.
Naunyn Schmiedebergs Arch Pharmacol ; 392(3): 287-297, 2019 03.
Article in English | MEDLINE | ID: mdl-30483860

ABSTRACT

Liver injury, one of the major side effects of diclofenac (DIC), plagues thousands of patients who treated with it. Although involvements of metabolic factors, oxidative stress, and mitochondrial injury have been characterized, the exact immunomolecular mechanism of the hepatotoxicity of DIC still remains ambiguous. In this study, we investigated the role of chemokine receptors CCR2 and CCR5 in this progression. Ccr2, Ccr5, and Tnfr1/2-deficient mice, as well as wild type littermates, were administrated DIC or vehicle for 24 h, receptively. Hepatic expression of CCR2, CCR5, and their ligands were upregulated after DIC treatment. DIC-induced liver injury was augmented in Ccr2+/+ mice than Ccr2-/- mice, a similar phenotype was observed in Ccr5-deficient mice. In addition, antagonists of CCR2 or CCR5 protected liver damage caused by diclofenac. Besides, the number of neutrophils present in the liver was gradually increased from 0 to 12 h after drug administration. However, the recruitment of neutrophils was dramatically lessened after blocking CCR2 or CCR5 signaling. Furthermore, TNF-α level in the liver was decreased in Ccr2-/- mice compared with Ccr2+/+ mice. Intriguingly, in line with this, TNF receptor 1 and 2 double knockout mice showed markedly attenuated hepatotoxicity of DIC. These suggested that CCR2 and CCR5 mediated hepatotoxicity induced by diclofenac, TNF-α was responsible, at least in part, for it, and the pharmacological inhibition of CCR2 or CCR5 might serve as a novel therapeutic approach for DIC-induced hepatotoxicity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Chemical and Drug Induced Liver Injury/immunology , Diclofenac/adverse effects , Receptors, CCR2/immunology , Receptors, CCR5/immunology , Animals , Chemical and Drug Induced Liver Injury/genetics , Cytochrome P-450 Enzyme System/genetics , Glucuronosyltransferase/genetics , Liver/drug effects , Liver/immunology , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Receptors, CCR2/genetics , Receptors, CCR5/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Biochem Biophys Res Commun ; 508(4): 1004-1010, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30551881

ABSTRACT

Sepsis is a serious condition with a high mortality rate worldwide. Granisetron is an anti-nausea drug for patients undergoing chemotherapy. Here we aimed to identify the novel effect of granisetron on sepsis-induced acute lung injury (ALI). Our results showed that mice treated with granisetron displayed less severe lung damage than controls. Granisetron administration reduced pulmonary neutrophil recruitment after CLP. Moreover, the expressions of Cxcl1 and Cxcl2 were diminished in the presence of granisetron in THP-1 macrophages after lipopolysaccharide exposure. Additionally, granisetron could inhibit the activation of p38 MAPK and NLRP3 inflammasome both in vivo and in vitro. Collectively, granisetron protects against sepsis-induced ALI by suppressing macrophage Cxcl1/Cxcl2 expression and neutrophil recruitment in the lung.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/microbiology , Granisetron/therapeutic use , Sepsis/drug therapy , Sepsis/microbiology , Acute Lung Injury/pathology , Animals , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Granisetron/pharmacology , Humans , Inflammasomes/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophil Infiltration/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Sepsis/pathology , THP-1 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Transl Med ; 16(1): 344, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30526624

ABSTRACT

BACKGROUND: L-Fucose (Fuc), a six-deoxy hexose monosaccharide, is present endogenously in humans and animals and has a wide range of biological functions. In the present study, we aimed to examine the effect of Fuc on obesity and hepatic steatosis in mice fed a high-fat diet (HFD). METHODS: C57BL/6 mice were fed a normal chow (NC) or HFD for 18 weeks to induce obesity and fatty liver. Fuc was administered intragastrically from the 8th week to the end of the experiment (18 weeks). RESULTS: Metagenomic analysis showed that HFD altered the genomic profile of gut microbiota in the mice; specifically, expression of alpha-L-fucosidase, the gene responsible for Fuc generation, was markedly reduced in the HFD group compared with that in the NC group. Fuc treatment decreased body weight gain, fat accumulation, and hepatic triglyceride elevation in HFD-fed mice. In addition, Fuc decreased the levels of endotoxin-producing bacteria of the Desulfovibrionaceae family and restored HFD-induced enteric dysbiosis at both compositional and functional levels. CONCLUSION: Our findings suggest that Fuc might be a novel strategy to treat HFD-induced obesity and fatty liver.


Subject(s)
Diet, High-Fat , Fucose/therapeutic use , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/complications , Obesity/drug therapy , Adiposity/drug effects , Animals , Cecum/drug effects , Cecum/microbiology , Dysbiosis/complications , Dysbiosis/microbiology , Dysbiosis/pathology , Feeding Behavior , Fucose/pharmacology , Gastrointestinal Microbiome/drug effects , Glucose Tolerance Test , Insulin , Metagenomics , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/microbiology , Obesity/microbiology , Weight Gain/drug effects
11.
Biochem Biophys Res Commun ; 501(1): 16-23, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29730287

ABSTRACT

The gut microbiota exhibit diurnal compositional and functional oscillations that influence the host homeostasis. However, the upstream factors that affect the microbial oscillations remain elusive. Here, we focused on the potential impact of light exposure, the main factor that affects the host circadian oscillation, on the diurnal oscillations of intestinal microflora to explore the upstream factor that governs the fluctuations of the gut microbes. The gut microbiota of the mice that were underwent regular light/dark (LD) cycles exhibited a robust rhythm at both compositional and functional level, in all parts of the intestine. Comparably, constant darkness (Dark-Dark, DD) led to the loss of the rhythmic oscillations in almost all parts of the intestine. Additionally, the abundance of Clostridia in DD conditions was dramatically enhanced in the small intestine. Our data indicated light exposure is the upstream factor that governs the regular diurnal fluctuations of gut microbiota in vivo.


Subject(s)
Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/radiation effects , Light , Photoperiod , Animals , Cecum/microbiology , Cecum/radiation effects , Clostridium/isolation & purification , Darkness , Gastrointestinal Microbiome/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/radiation effects , Intestine, Small/microbiology , Intestine, Small/radiation effects , Male , Mice , Mice, Inbred BALB C
12.
Sensors (Basel) ; 14(10): 18878-85, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25310474

ABSTRACT

As a network transmission protocol, Networked Transport of RTCM via Internet Protocol (NTRIP) is widely used in GPS and Global Orbiting Navigational Satellite System (GLONASS) Augmentation systems, such as Continuous Operational Reference System (CORS), Wide Area Augmentation System (WAAS) and Satellite Based Augmentation Systems (SBAS). With the deployment of BeiDou Navigation Satellite system(BDS) to serve the Asia-Pacific region, there are increasing needs for ground monitoring of the BeiDou Navigation Satellite system and the development of the high-precision real-time BeiDou products. This paper aims to optimize the decoding algorithm of NTRIP Client data streams and the user authentication strategies of the NTRIP Caster based on NTRIP. The proposed method greatly enhances the handling efficiency and significantly reduces the data transmission delay compared with the Federal Agency for Cartography and Geodesy (BKG) NTRIP. Meanwhile, a transcoding method is proposed to facilitate the data transformation from the BINary EXchange (BINEX) format to the RTCM format. The transformation scheme thus solves the problem of handing real-time data streams from Trimble receivers in the BeiDou Navigation Satellite System indigenously developed by China.

SELECTION OF CITATIONS
SEARCH DETAIL