Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 728: 150340, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38968770

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a prevalent malignancy and poses a significant clinical challenge. Piperine, an alkaloid molecule extracted from Piper nigrum and Piper longum, has emerged as a promising anticancer agent. However, the molecular mechanisms of piperine' antitumor effects in CRC need to be further elucidated. METHODS: Human colorectal cancer cells were treated with piperine in vitro. CCK-8 and clone formation assays were adopted to detect cell viability. The accumulation of autophagosomes was assessed by Western blotting and immunofluorescence. Apoptosis and reactive oxygen species (ROS) levels were analyzed by flow. In vivo, a xenograft tumor mouse model was constructed using CT26 cells. RESULTS: Piperine inhibited CRC cell viability and suppressed tumor weight and volume in a mouse model. Additionally, piperine treatment induced the accumulation of autophagosomes in CRC cells. This effect was attributed to the inhibition of the AKT/mTOR pathway and the accumulation of ROS. activation of AKT or clearance of ROS attenuated piperine-mediated tumor suppression. CONCLUSION: This study shows that piperine induces autophagy-dependent cell death in CRC cells by increasing ROS production and inhibiting Akt/mTOR signaling.

2.
Front Med (Lausanne) ; 11: 1368977, 2024.
Article in English | MEDLINE | ID: mdl-38947241

ABSTRACT

Intestinal fibrosis is a common complication of chronic intestinal diseases with the characteristics of fibroblast proliferation and extracellular matrix deposition after chronic inflammation, leading to lumen narrowing, structural and functional damage to the intestines, and life inconvenience for the patients. However, anti-inflammatory drugs are currently generally not effective in overcoming intestinal fibrosis making surgery the main treatment method. The development of intestinal fibrosis is a slow process and its onset may be the result of the combined action of inflammatory cells, local cytokines, and intestinal stromal cells. The aim of this study is to elucidate the pathogenesis [e.g., extracellular matrix (ECM), cytokines and chemokines, epithelial-mesenchymal transition (EMT), differentiation of fibroblast to myofibroblast and intestinal microbiota] underlying the development of intestinal fibrosis and to explore therapeutic advances (such as regulating ECM, cytokines, chemokines, EMT, differentiation of fibroblast to myofibroblast and targeting TGF-ß) based on the pathogenesis in order to gain new insights into the prevention and treatment of intestinal fibrosis.

3.
Med Oncol ; 39(12): 211, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36175702

ABSTRACT

Colorectal cancer (CRC) is a cancer with a high morbidity and mortality worldwide. Hence, developing new therapeutic drugs for CRC is very important. Moxidectin (MOX) has shown good anti-glioblastoma effect both in vitro and in vivo. This study aimed to elucidate the anti-CRC effect of MOX and its potential mechanism by investigating the influence of MOX on the viability, apoptosis, necrosis and autophagy of colorectal cancer cells (HCT15 and SW620) and its underlying mechanisms. It was found that MOX can induce autophagy arrest, promote autophagy initiation, inhibit autophagic flux and cell proliferation, simultaneously PI3K-Akt-mTOR signaling pathway and microtubule acetylation. Furthermore, MOX suppressed the growth of xenograft tumors, which was consistent with the in vitro results.


Subject(s)
Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , Autophagy , Colorectal Neoplasms/drug therapy , Humans , Macrolides , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases
4.
Theranostics ; 12(10): 4802-4817, 2022.
Article in English | MEDLINE | ID: mdl-35832094

ABSTRACT

Background: Chemoresistance to 5-fluorouracil (5-FU) is a major barrier to influence the treatment efficiency of colorectal cancer (CRC) patients, while the precise molecular mechanisms underlying 5-FU resistance remain to be fully elucidated. Methods: The metabolic profiles including ATP generation, glucose consumption, lactate generation, and oxygen consumption rate (OCR) in 5-FU resistant CRC cells were compared with those in their parental cells. Subsequently, a series of in vitro and in vivo experiments were carried out to investigate the mechanisms responsible for metabolic reprogramming of 5-FU resistant CRC cells. Results: We found that 5-FU resistant CRC cells showed increased levels of ATP generation, glucose consumption, lactate generation, and OCR as compared with those in their parental cells. Further, increased levels of mRNA N6-methyladenosine (m6A) and methyltransferase-like 3 (METTL3) were observed in 5-FU resistant CRC cells. Inhibition or knockdown of METTL3 can suppress glycolysis and restore chemosensitivity of 5-FU resistant CRC cells. Mechanistically, METTL3 enhances the expression of LDHA, which catalyzes the conversion of pyruvate to lactate, to trigger glycolysis and 5-FU resistance. METTL3 can increase the transcription of LDHA via stabilizing mRNA of hypoxia-inducible factor (HIF-1α), further, METTL3 also triggers the translation of LDHA mRNA via methylation of its CDS region and recruitment of YTH domain-containing family protein 1 (YTHDF1). Targeted inhibition of METTL3/LDHA axis can significantly increase the in vitro and in vivo 5-FU sensitivity of CRC cells. Conclusion: Our study indicates that METTL3/LDHA axis-induced glucose metabolism is a potential therapy target to overcome 5-FU resistance in CRC cells.


Subject(s)
Adenosine , Colorectal Neoplasms , Fluorouracil , L-Lactate Dehydrogenase , Adenosine/analogs & derivatives , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Glucose/metabolism , HCT116 Cells , Humans , L-Lactate Dehydrogenase/biosynthesis , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactates/metabolism , Methyltransferases/genetics , RNA, Messenger
5.
Int J Biol Sci ; 18(6): 2540-2552, 2022.
Article in English | MEDLINE | ID: mdl-35414768

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies worldwide. Metastasis is a major cause of CRC recurrence and mortality. Several antibiotic drugs have been reported to exert potential anticancer activities, however, whether and how the tetracycline antibiotic minocycline exhibit tumor suppressive effect on CRC remains unknown. Here, we found that minocycline markedly inhibits the epithelial-mesenchymal transition (EMT) process and metastasis of CRC cells both in vitro and in vivo. Using chemical proteomics screening combined with docking analysis and site-directed mutagenesis, we identified LYN as a direct bind target of minocycline, and Ala255 of LYN is required for minocycline binding. Mechanistically, minocycline binding inactivates LYN, leading to STAT3 inactivation and EMT suppression, thereby inhibits CRC metastasis. Tissue microarray analysis further confirmed the clinical relevance of LYN-STAT3 axis in the EMT and progression of CRC. In addition to CRC, minocycline also significantly prevents EMT process and inhibits the metastasis of several other cancer types. Our findings elucidate the mechanism of action of minocycline for the inhibition of CRC metastasis by LYN binding, and suggest that repurposing minocycline may represent a promising strategy for the treatment of advanced CRC and other cancer types.


Subject(s)
Colorectal Neoplasms , Minocycline , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Minocycline/pharmacology , Minocycline/therapeutic use , Neoplasm Metastasis , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
6.
Aging (Albany NY) ; 12(23): 24424-24440, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33291076

ABSTRACT

Colorectal cancer (CRC) is the third most common type of cancer worldwide. Metastasis and chemoresistance are regarded as the two leading causes of treatment failure and high mortality in CRC. Forkhead Box M1 (FOXM1) has been involved in malignant behaviors of cancer. However, the role and mechanism of FOXM1 in simultaneously regulating metastasis and chemoresistance of CRC remain poorly understood. Here, we found that FOXM1 was overexpressed in oxaliplatin- and vincristine-resistant CRC cells (HCT-8/L-OHP and HCT-8/VCR) with enhanced metastatic potential, compared with HCT-8 cells. FOXM1 overexpression increased migration, invasion and drug-resistance to oxaliplatin and vincristine in HCT-8 cells, while FOXM1 knockdown using shFOXM1 impaired metastasis and drug-resistance in HCT-8/L-OHP and HCT-8/VCR cells. Moreover, FOXM1 up-regulated Snail to trigger epithelial-mesenchymal transition-like molecular changes and multidrug-resistance protein P-gp expression, while silencing Snail inhibited FOXM1-induced metastasis and drug-resistance. We further identified that disheveled-2 (DVL2) was crucial for FOXM1-induced Snail expression, metastasis and chemoresistance. Furthermore, FOXM1 bound to DVL2, and enhanced nuclear translocation of DVL2 and DVL2-mediated transcriptional activity of Wnt/ß-catenin known to induce Snail expression. In conclusion, FOXM1/DVL2/Snail axis triggered aggressiveness of CRC. Blocking FOXM1/DVL2/Snail pathway simultaneously inhibited metastasis and chemoresistance in CRC cells, providing a new strategy for successful CRC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Dishevelled Proteins/metabolism , Drug Resistance, Neoplasm , Forkhead Box Protein M1/metabolism , Oxaliplatin/pharmacology , Snail Family Transcription Factors/metabolism , Vincristine/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dishevelled Proteins/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Snail Family Transcription Factors/genetics , Wnt Signaling Pathway
7.
Cancer Cell Int ; 20: 205, 2020.
Article in English | MEDLINE | ID: mdl-32514250

ABSTRACT

BACKGROUND: Inflammation is one of a main reason for colon cancer progression and poor prognosis. The high-mobility group box-1 (HMGB1) and glutathione peroxidase 4 (GPX4) are responsible for inflammation, but the relationship between HMGB1 and GPX4 remains unknown about inflammation in colon cancer. METHODS: RT-qPCR was carried out to investigate the expression of IL1ß, IL6 and TNFα in colon cancer cells stimulated with LPS or siHMGB1. To observe the relationship between HMGB1, GPX4 and inflammation or ROS, Western blot assays were adopted. Pull-down, CoIP and immunohistochemistry assays were performed to further investigate the molecular mechanisms of HMGB1 and GPX4 in colon cancer. RESULTS: We report that HMGB1 mediates lipopolysaccharide (LPS)-induced inflammation in colon cancer cells. Mechanistically, acetylated HMGB1 interacts with GPX4, negatively regulating GPX4 activity. Furthermore, by utilizing siHMGB1 and its inhibitor, our discoveries demonstrate that HMGB1 knockdown can inhibit inflammation and reactive oxygen species (ROS) accumulation via NF-kB. CONCLUSION: Collectively, our findings first demonstrate that acetylated HMGB1 can interact with GPX4, leading to inflammation, and providing therapeutic strategies targeting HMGB1 and GPX4 for colon cancer.

8.
Cell Prolif ; 51(3): e12427, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29292543

ABSTRACT

OBJECTIVES: Alzheimer's disease (AD) is one of the most prevalent brain diseases among the elderly, majority of which is caused by abnormal deposition of amyloid beta-peptide (Aß). Galantamine, currently the first-line drug in treatment of AD, has been shown to diminish Aß-induced neurotoxicity and exert favourable neuroprotective effects, but the detail mechanisms remain unclear. MATERIALS AND METHODS: Effects of galantamine on Aß-induced cytotoxicity were checked by MTT, clone formation and apoptosis assays. The protein variations and reactive oxygen species (ROS) production were measured by western blotting analysis and dichloro-dihydro-fluorescein diacetate assay, respectively. RESULTS: Galantamine reversed Aß-induced cell growth inhibition and apoptosis in neuron cells PC12. Aß activated the entire autophagy flux and accumulation of autophagosomes, and the inhibition of autophagy decreased the protein level of cleaved-caspase-3 and Aß-induced cytotoxicity. Meanwhile, galantamine suppressed Aß-mediated autophagy flux and accumulation of autophagosomes. Moreover, Aß upregulated ROS accumulation, while ROS scavengers N-acetyl-l-cysteine impaired Aß-mediated autophagy. Further investigation showed that galantamine downregulated NOX4 expression to inhibit Aß-mediated ROS accumulation and autophagy. CONCLUSIONS: Galantamine inhibits Aß-induced cytostatic autophagy through decreasing ROS accumulation, providing new insights into deep understanding of AD progression and molecular basis of galantamine in neuroprotection.


Subject(s)
Amyloid beta-Peptides/physiology , Autophagy/drug effects , Cytostatic Agents/pharmacology , Galantamine/pharmacology , Peptide Fragments/physiology , Reactive Oxygen Species/metabolism , Alzheimer Disease/drug therapy , Animals , Apoptosis , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , NADPH Oxidase 4/metabolism , PC12 Cells , Rats
9.
J Cell Mol Med ; 22(1): 89-100, 2018 01.
Article in English | MEDLINE | ID: mdl-28799229

ABSTRACT

Recently, the dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin, a major anti-hyperglycaemic agent, has received substantial attention as a therapeutic target for cardiovascular diseases via enhancing the number of circulating endothelial progenitor cells (EPCs). However, the direct effects of sitagliptin on EPC function remain elusive. In this study, we evaluated the proangiogenic effects of sitagliptin on a diabetic hind limb ischaemia (HLI) model in vivo and on EPC culture in vitro. Treatment of db/db mice with sitagliptin (Januvia) after HLI surgery efficiently enhanced ischaemic angiogenesis and blood perfusion, which was accompanied by significant increases in circulating EPC numbers. EPCs derived from the bone marrow of normal mice were treated with high glucose to mimic diabetic hyperglycaemia. We found that high glucose treatment induced EPC apoptosis and tube formation impairment, which were significantly prevented by sitagliptin pretreatment. A mechanistic study found that high glucose treatment of EPCs induced dramatic increases in oxidative stress and apoptosis; pretreatment of EPCs with sitagliptin significantly attenuated high glucose-induced apoptosis, tube formation impairment and oxidative stress. Furthermore, we found that sitagliptin restored the basal autophagy of EPCs that was impaired by high glucose via activating the AMP-activated protein kinase/unc-51-like autophagy activating kinase 1 signalling pathway, although an autophagy inhibitor abolished the protective effects of sitagliptin on EPCs. Altogether, the results indicate that sitagliptin-induced preservation of EPC angiogenic function results in an improvement of diabetic ischaemia angiogenesis and blood perfusion, which are most likely mediated by sitagliptin-induced prevention of EPC apoptosis via augmenting autophagy.


Subject(s)
Autophagy , Diabetes Mellitus, Type 2/drug therapy , Endothelial Progenitor Cells/pathology , Ischemia/drug therapy , Neovascularization, Physiologic , Sitagliptin Phosphate/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Survival/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Glucose/toxicity , Hindlimb/blood supply , Ischemia/complications , Ischemia/pathology , Male , Mice , Neovascularization, Physiologic/drug effects , Perfusion , Reactive Oxygen Species/metabolism , Regional Blood Flow/drug effects , Signal Transduction , Sitagliptin Phosphate/pharmacology
10.
Circ Res ; 120(5): e7-e23, 2017 Mar 03.
Article in English | MEDLINE | ID: mdl-28137917

ABSTRACT

RATIONALE: Endothelial progenitor cells (EPCs) respond to stromal cell-derived factor 1 (SDF-1) through chemokine receptors CXCR7 and CXCR4. Whether SDF-1 receptors involves in diabetes mellitus-induced EPCs dysfunction remains unknown. OBJECTIVE: To determine the role of SDF-1 receptors in diabetic EPCs dysfunction. METHODS AND RESULTS: CXCR7 expression, but not CXCR4 was reduced in EPCs from db/db mice, which coincided with impaired tube formation. Knockdown of CXCR7 impaired tube formation of EPCs from normal mice, whereas upregulation of CXCR7 rescued angiogenic function of EPCs from db/db mice. In normal EPCs treated with oxidized low-density lipoprotein or high glucose also reduced CXCR7 expression, impaired tube formation, and increased oxidative stress and apoptosis. The damaging effects of oxidized low-density lipoprotein or high glucose were markedly reduced by SDF-1 pretreatment in EPCs transduced with CXCR7 lentivirus but not in EPCs transduced with control lentivirus. Most importantly, EPCs transduced with CXCR7 lentivirus were superior to EPCs transduced with control lentivirus for therapy of ischemic limbs in db/db mice. Mechanistic studies demonstrated that oxidized low-density lipoprotein or high glucose inhibited protein kinase B and glycogen synthase kinase-3ß phosphorylation, nuclear export of Fyn and nuclear localization of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), blunting Nrf2 downstream target genes heme oxygenase-1, NAD(P)H dehydrogenase (quinone 1) and catalase, and inducing an increase in EPC oxidative stress. This destructive cascade was blocked by SDF-1 treatment in EPCs transduced with CXCR7 lentivirus. Furthermore, inhibition of phosphatidylinositol 3-kinase/protein kinase B prevented SDF-1/CXCR7-mediated Nrf2 activation and blocked angiogenic repair. Moreover, Nrf2 knockdown almost completely abolished the protective effects of SDF-1/CXCR7 on EPC function in vitro and in vivo. CONCLUSIONS: Elevated expression of CXCR7 enhances EPC resistance to diabetes mellitus-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia. The benefits of CXCR7 are mediated predominantly by a protein kinase B/glycogen synthase kinase-3ß/Fyn pathway via increased activity of Nrf2.


Subject(s)
Diabetes Mellitus/metabolism , Endothelial Progenitor Cells/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Ischemia/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, CXCR/biosynthesis , Animals , Cells, Cultured , Diabetes Mellitus/pathology , Gene Knockdown Techniques , HEK293 Cells , Hindlimb/blood supply , Hindlimb/metabolism , Hindlimb/pathology , Humans , Ischemia/pathology , Male , Mice , Mice, Transgenic , NF-E2-Related Factor 2/metabolism , Neovascularization, Physiologic/physiology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism
11.
Toxicol In Vitro ; 30(1 Pt B): 521-8, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26363202

ABSTRACT

The effects and mechanisms of bisphenol A (BPA) on the development of breast cancer are still not well illustrated. The present study revealed that nanomolar BPA significantly promoted the proliferation of both estrogen receptor (ER) positive (MCF-7) and negative (SkBr3) breast cancer cells, which was confirmed by up regulation of proliferating cell nuclear antigen (PCNA) and Bcl-2. Neither ERα nor G-protein-coupled estrogen receptor (GPER) mediated this effect of BPA because their inhibitors had no effect on the BPA induced cell proliferation. However, silencing of estrogen related receptor gamma (ERRγ) by its specific siRNA significantly abolished BPA induced proliferation of breast cancer cells, while si-ERRα had no similar effect. Moreover, nanomolar BPA up regulated the mRNA and protein levels of ERRγ and triggered its nuclear translocation via a time dependent manner. Further studies revealed that 10(-8)M BPA obviously increased the phosphorylation of ERK1/2, while had no similar effect on the phosphorylation of JNK and p38 MAPK. Further, PD 98059, the inhibitor of ERK1/2, significantly abolished the BPA induced up regulation of ERRγ and proliferation of breast cancer cells. Collectively, our results revealed that nanomolar BPA can trigger the proliferation of breast cancer cells via ERK1/2/ERRγ signals. Given that nanomolar BPA has been widely detected in human tissues, the clinical relevance of BPA and breast cancer progression should be further investigated.


Subject(s)
Benzhydryl Compounds/pharmacology , Breast Neoplasms/pathology , MAP Kinase Signaling System/physiology , Phenols/pharmacology , Receptors, Estrogen/physiology , Active Transport, Cell Nucleus , Cell Proliferation/drug effects , Estrogen Receptor alpha/physiology , Female , Humans , MCF-7 Cells , Receptors, G-Protein-Coupled/physiology , Signal Transduction
12.
Cell Prolif ; 48(2): 249-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25643607

ABSTRACT

OBJECTIVES: Expression of dishevelled-1 (DVL1) has recently been linked to cancer progression, however, its role in resistance to cancer therapy is unclear. In this study, we aimed to explore the function of DVL1 in paclitaxel-resistant human ovarian cancer cells. MATERIALS AND METHODS: The MTT assay was used to assess effects of DVL1 silencing on sensitivity of cells that were otherwise resistant to paclitaxel (Taxol). Western blotting and immunofluorescence staining were used to examine effects of DVL1 on AKT/GSK-3ß/ß-catenin signalling. RESULTS: Dishevelled-1 was found to be over-expressed in a paclitaxel-resistant cell line derived from human ovarian cancer cell line A2780 (A2780/Taxol line) as well as parental A2780 cells. Down-regulation of DVL1 (using the inhibitor 3289-8625 or siRNA (siDVL1) against DVL1) sensitized A2780/Taxol cells to paclitaxel. Over-expression of DVL1 in A2780 cells increased protein levels of P-gp, BCRP and Bcl-2, which are known targets of ß-catenin. Silencing DVL1 in A2780/Taxol cells also reduced levels of these proteins, and led to accumulation of ß-catenin. In addition, DVL1 aberrantly activated AKT/GSK-3ß/ß-catenin signalling. Inactivation of AKT signalling attenuated DVL1-mediated inhibition of GSK-3ß and accumulation of ß-catenin, in both A2780 and A2780/Taxol cells. CONCLUSIONS: Taken together, these results suggest that silencing DVL1 sensitized A2780/Taxol cells to paclitaxel, by down-regulating AKT/GSK-3ß/ß-catenin signalling, providing a novel strategy for chemosensitization of ovarian cancer to paclitaxel-induced cytotoxicity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , Phosphoproteins/genetics , Wnt Signaling Pathway/genetics , Active Transport, Cell Nucleus/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/biosynthesis , Cell Line, Tumor , Dishevelled Proteins , Enzyme Activation/genetics , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/biosynthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction , beta Catenin/metabolism
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(7): 994-8, 2013 Jul.
Article in Chinese | MEDLINE | ID: mdl-23895839

ABSTRACT

OBJECTIVE: To explore the effects of CXCR7 knock-down by CXCR7-shRNA lentiviral vector on the proliferation and invasion of human hepatoma carcinoma cells in vitro. METHODS: CXCR7-shRNA lentiviral vector was transfected into hepatocellular carcinoma HCCLM3 cells. The changes in mRNA and protein expression of CXCR7 in the transfected cells were investigated using real-time PCR and Western blotting, respectively, and MTT assay was employed to assess the cell proliferation changes. In vitro adhesion assay and transwell chamber test were used to observe the adhesion and invasiveness of HCCLM3 cells, respectively. RESULTS: Transfection of HCCLM3 cells with CXCR7-shRNA lentiviral vector resulted in a significantly decreased expression of CXCR7 at both mRNA and protein levels (P<0.01) and obvious suppression of the cell proliferative activity (P<0.05). CXCR7-shRNA also significantly suppressed the invasiveness and adhesion of HCCLM3 cells (P<0.01). CONCLUSION: CXCR7 knock-down can significantly inhibit the proliferation and invasiveness of human hepatoma carcinoma cells in vitro, suggesting the value of CXCR7 as a potential target for hepatoma carcinoma therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , RNA, Small Interfering , Receptors, CXCR/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Genetic Vectors , Humans , Lentivirus/genetics , Liver Neoplasms/genetics , RNA, Small Interfering/genetics , Transfection
14.
Zhonghua Nan Ke Xue ; 17(6): 546-52, 2011 Jun.
Article in Chinese | MEDLINE | ID: mdl-21735657

ABSTRACT

OBJECTIVE: To evaluate the association of gr/gr deletion in the AZFc region of Y chromosome with idiopathic male infertility using Meta-analysis. METHODS: All relevant case-control studies addressing the relationship between gr/gr deletion and idiopathic male infertility were identified from PubMed, VIP and CNKI (from January 2003 to August 2010). Statistical analyses were performed with the RevMan4. 2 software. RESULTS: Twenty eligible articles were selected in this study, including 5 246 cases of idiopathic infertility and 4 380 controls. The integrated data from the 20 studies revealed a significantly higher frequency of gr/gr deletion in the patients than in the controls, with an odds ratio (OR) of 1.63 (95% CI: 1.23 -2.44) (P = 0.002). However, when the Meta-analysis was limited to 16 studies with stricter case and control selection criteria, the overall OR increased to 1.84 (95% CI: 1.47 - 2.29) (P < 0.000 01). Thirteen studies showed that oligozoospermia patients had a significantly higher frequency of gr/gr deletion than controls (OR = 2.12, 95% CI: 1.61 - 2.80) (P < 0.000 01). Eight studies showed a significant association between the gr/gr deletion subtype without DAZ1/DAZ2 gene copies and spermatogenic impairment (OR = 1.83, 95% CI: 1.31 - 2.55) (P = 0.000 4), but no statistically significant differences were found in the frequency distribution of the gr/gr deletion subtype missing DAZ3/DAZ4 gene copies between the patients and controls (OR = 1.43, 95% CI: 0.97 -2.11) (P = 0.07). CONCLUSION: The present data suggest that gr/gr deletion may be one of the risk factors of male infertility.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Y , Infertility, Male/genetics , Humans , Male
15.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 24(5): 1133-6, 1166, 2007 Oct.
Article in Chinese | MEDLINE | ID: mdl-18027712

ABSTRACT

Hypoxia-inducible transcription factor-1alpha (HIF-1alpha) is a "master" gene in the cell hypoxia response network. HIF-1alpha plays the key role on VEGF-dependent angiogenesis during tumor development. In this study, the siRNA expressive vector targeting HIF-1alpha was constructed. HepG2 cells were tranfected with an HIF-1alpha siRNA vector and exposed to a normoxic or hypoxic environment. mRNA and protein levels of HIF-1alpha and VEGF were detected by RT-PCR and Western blot at different cell culture environment. The mRNA and protein expression of HIF-1alpha was inhibited by RNA interference. The obvious decreases of mRNA and protein of VEGF was induced by HIF-1alpha gene knock-down. Our study indicated that HIF-1alpha regulated the expression of VEGF and hence promoted angiogenesis in liver cancer under hypoxic environment. The expression of VEGF was downregulated via HIF-1alpha gene knock-down. These results suggest that HIF-1alpha may be used as a good target gene for anti-angiogenesis in liver tumors.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , RNA Interference , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Down-Regulation , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , RNA, Messenger/metabolism
16.
BMC Cell Biol ; 8: 1, 2007 Jan 16.
Article in English | MEDLINE | ID: mdl-17224075

ABSTRACT

BACKGROUND: Connective tissue growth factor (CTGF) is a potent profibrotic factor, which is implicated in fibroblast proliferation, angiogenesis and extracellular matrix (ECM) synthesis. It is a downstream mediator of some of the effects of transforming growth factor beta (TGFbeta) and is potentially induced by hyperglycemia in human renal mesangial cells. However, whether high glucose could induce the CTGF expression in vascular smooth muscle cells (VSMCs) remains unknown. Therefore, this study was designed to test whether high glucose could regulate CTGF expression in human VSMC. The effect of modulating CTGF expression on VSMC proliferation and migration was further investigated. RESULTS: Expression of CTGF mRNA was up-regulated as early as 6 hours in cultured human VSMCs after exposed to high glucose condition, followed by ECM components (collagen type I and fibronectin) accumulation. The upregulation of CTGF mRNA appears to be TGFbeta-dependent since anti-TGFbeta antibody blocks the effect of high glucose on CTGF gene expression. A small interference RNA (siRNA) targeting CTGF mRNA (CTGF-siRNA) effectively suppressed CTGF up-regulation stimulated by high glucose up to 79% inhibition. As a consequence of decreased expression of CTGF gene, the deposition of ECM proteins in the VSMC was also declined. Moreover, CTGF-siRNA expressing vector partially inhibited the high glucose-induced VSMC proliferation and migration. CONCLUSION: Our data suggest that in the development of macrovascular complications in diabetes, CTGF might be an important factor involved in the patho-physiological responses to high glucose in human VSMCs. In addition, the modulatory effects of CTGF-siRNA during this process suggest that specific targeting CTGF by RNA interference could be useful in preventing intimal hyperplasia in diabetic macrovascular complications.


Subject(s)
Glucose/pharmacology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Up-Regulation , Cell Movement/drug effects , Cell Proliferation/drug effects , Connective Tissue Growth Factor , Extracellular Matrix/metabolism , Glucose/metabolism , Humans , Myocytes, Smooth Muscle/cytology , RNA Interference , Transforming Growth Factor beta/physiology , Umbilical Veins/cytology
17.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 23(3): 625-9, 2006 Jun.
Article in Chinese | MEDLINE | ID: mdl-16856403

ABSTRACT

Survivin is a member of inhibitor of apoptosis (IAP) family which has strong ability of antiapoptosis. Survivin is prominently expressed in transformed cell lines and in all the most common human cancers in vivo, but it is undetectable in terminally differentiated adult tissues. We designed a human survivin antisense oligonucleotides. RT-PCR and Western blot clearly showed that survivin mRNA and protein were decreased by this antisense oligonucleotide. The inhibition of SMMC-7721 cell proliferation was demonstrated by MTT assay. The inhibition rate (43.28% +/- 3.65%) was much higher than the inhibition rate (6.76% +/- 0.92%) of the control group. The apoptosis rate (33.68% +/- 2.89%) obtained by transfecting cells with the use of antisense oligonucleotide was higher than the apoptosis rate (14.12% +/- 1.24%) of the control group. And the result of TUNEL revealed that, when SMMC-7721 cells were exposed to homoharringtoine at low concentration and were transfected by antisense oligonucleotide, the apoptosis rate of the cells was significantly higher than that of other control group. These results indicate that antisense oligonucleotide may have the potential for selective tumor therapy in future.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Oligonucleotides, Antisense/pharmacology , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...