Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Article in English | MEDLINE | ID: mdl-38664244

ABSTRACT

Acute lung injury (ALI) is a common disease with complex pathogenesis. However, the treatment is mainly symptomatic with limited clinical options. Asiaticoside (AS), a Chinese herbal extract, has protective effects against LPS-induced ALI in mice and inhibits nitric oxide and prostaglandin E2 synthesis; however, the specific mechanism of AS in the prevention and treatment of LPS-induced ALI needs further study. Sema4D/CD72 pathway, mitochondrial dysfunction, and miRNA-21 are closely associated with inflammation. Therefore, the present study aimed to explore whether AS exerts its therapeutic effect on ALI by influencing Sema4D/CD72 pathway and mitochondrial dysfunction, restoring the balance of inflammatory factors, and influencing miRNA-21 expression. Cell and animal experiments were performed to investigate the effect of AS on ALI. Lipopolysaccharide (LPS) was used to establish the ALI model. CCK8 and flow cytometry were used to detect the cell viability and apoptosis rate. HE staining and wet-to-dry weight ratio (W/D) of lung tissue were determined. The expressions of Sema4D, CD72, NF-κB p65, Bax, Bcl2, and caspase 3 in RAW264.7 cells and lung tissues were detected by western blot, and the levels of IL-10 and IL-1ß induced by LPS in supernatant of RAW264.7 cells and BALF were measured by ELISA. And the expression of miRNA-21 in cells and lung tissues was detected by fluorescence quantitative PCR. The result shows that AS treatment suppressed LPS-induced cell damage and lung injury in mice. AS treatment could alleviate the pathological changes such as inflammatory infiltration and histopathological changes in the lungs caused by LPS, and reduce the ratio of W/D. AS significantly alleviated the decrease of mitochondrial membrane potential induced by LPS, inhibited the increase of ROS production, and reduced the expression of mitochondrial fission proteins Drp1 and Fis1. The high-dose AS group significantly downregulated the expression of Sema4D, CD72, phosphorylated NF-κB p65, and apoptosis-related proteins, decreased the pro-inflammatory factor IL-1ß, and enhanced the level of anti-inflammatory factor IL-10. In addition, AS promoted miRNA-21 expression. These effects inhibited apoptosis and restored the balance between anti- and pro-inflammatory factors. This represents the inaugural report elucidating the mechanism by which AS inhibits the Sema4D/CD72 signaling pathway. These findings offer novel insights into the potential application of AS in both preventing and treating ALI.

3.
Biomed Chromatogr ; 35(2): e4968, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32881002

ABSTRACT

In this study, a simple and sensitive UHPLC-ESI-MS/MS method was established for the determination of LXH254 in rat plasma. The developed method was validated according to the Food and Drug administration guidelines. After extraction using ethyl acetate, the sample was separated on an ACQUITY BEH C18 column. The mobile phase consisted of 2 mM ammonium acetate containing 0.1% formic acid and acetonitrile as the mobile phase with gradient elution. The flow rate was 0.3 mL/min. A TSQ triple quadrupole mass spectrometer operated in positive-ion mode was used for mass detection, with multiple reaction monitoring transitions of m/z 503.3 > 459.1 and m/z 435.3 > 367.1 for LXH254 and olaparib (internal standard), respectively. An excellent linearity was achieved in the concentration range of 0.1-1000 ng/mL, with correlation coefficient >0.998. The mean recovery was more than 78.55%. Inter- and intra-day precision (percentage of relative standard deviation) did not exceed 12.87%, and accuracy was in the range of -2.50 to 13.50%. LXH254 was demonstrated to be stable under the tested storage conditions. The validated UHPLC-MS/MS method was further applied to the pharmacokinetic study of LXH254 in rat plasma after oral (2, 5, and 15 mg/kg) and intravenous (2 mg/kg) administrations. The pharmacokinetic study revealed that LXH254 showed low clearance, moderate bioavailability (~30%), and linear pharmacokinetic profile over the oral dose range of 2-15 mg/kg. To the best of our knowledge, this is the first report on the method development and validation of the determination of LXH254 and its application to pharmacokinetic study.


Subject(s)
Chromatography, High Pressure Liquid/methods , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Tandem Mass Spectrometry/methods , raf Kinases/antagonists & inhibitors , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Drug Stability , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Linear Models , Male , Niacinamide/analogs & derivatives , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
4.
Ann Palliat Med ; 9(4): 2079-2089, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32692224

ABSTRACT

BACKGROUND: Despite advances in therapeutic strategies and critical care management, septic encephalopathy (SE) is still a leading cause of infection-associated death in intensive care units (ICUs). Vitexin, a flavonoids compound, exerts and anti-inflammatory effect through inhibition of proinflammatory cytokines and signaling pathways. This study aimed to explore the anti-inflammatory effects of vitexin in SE and the underlying mechanisms. METHODS: An SE-inducedC57BL/6 mouse model was established via cecal ligation and puncture (CLP). Western blotting was performed to evaluate the protein expression levels of Chemokine (C-X-C motif) ligand 1 (CXCL1), fractalkine (CX3CL1), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), E-selectin, NF-κB p65, p-NF-κB p65, and tumor necrosis factor-α (TNF-α). Flow cytometry was used to detect the expressions ofCD11a/CD18, CD11b/CD18, ICAM-1, and adherent leukocyte. The expression of ICAM-1 was detected by immunohistochemistry. An enzyme-linked immunosorbent assay was performed to evaluate the expression of monocyte chemotactic protein-1 (MCP-1), Interleukin (IL)-6, IL-8, and IL-10. RESULTS: In this study, we found that vitexin significantly downregulated the expression of brain endothelial chemokines CXCL1 and CX3CL1 in CLP mice, exerting a potential anti-inflammatory against SE. Our data also showed that vitexin alleviated SE primarily by relying on reducing leukocyte-endothelial adhesion via the mediation of adhesion molecules. Moreover, vitexin suppressed the expression of proinflammatory cytokines, such as MCP-1, IL-6, IL-8, TNF-α, and NF-κB p65, in the CLP mice, while the expression of the anti-inflammatory cytokine IL-10 was elevated. CONCLUSIONS: Overall, our study demonstrated the protective effect vitexin exerts in SE by reducing leukocyte-endothelial adhesion and inflammatory response. These findings offer a molecular basis for the potential application of vitexin in the treatment of SE and other inflammatory-mediated and immunemediated disorders.


Subject(s)
Apigenin , Brain Diseases , Intercellular Adhesion Molecule-1 , Animals , Apigenin/pharmacology , Brain Diseases/immunology , Brain Diseases/therapy , Cell Adhesion , Endothelium , Inflammation , Leukocytes , Mice , Tumor Necrosis Factor-alpha , Vascular Cell Adhesion Molecule-1
5.
J Cancer ; 9(22): 4263-4270, 2018.
Article in English | MEDLINE | ID: mdl-30519328

ABSTRACT

The transcriptional coactivator with the PDZ-binding motif (TAZ) has been associated with different types of cancer. In this study, we examined the TAZ protein expression and cellular localization in 194 cases of human cervical squamous cell carcinoma (SCC). We observed that a normal cervix is characterized by higher expression levels of both nuclear and cytosolic TAZ compared to cervical SCC. Lower membranous and cytosolic TAZ expression levels are associated with lymph node involvement. We observed that TAZ expression levels are associated with ß1 integrin and Src in SCC and cell lines derived from human cervical cancers. Of note, knock down of TAZ increased the expression of ß1 integrin and Src in both normal and human cervical cancer cells. Our data indicate that the expression and cellular localization of TAZ are inversely associated with the development and progression of cervical SCC, and TAZ-mediated transcription may be involved in the activation of the integrin-Src signalling pathway.

6.
J Exp Clin Cancer Res ; 37(1): 171, 2018 Jul 27.
Article in English | MEDLINE | ID: mdl-30049286

ABSTRACT

BACKGROUND: Currently, resistance to tyrosine kinase inhibitors, such as gefitinib, has become a major obstacle in improving the clinical outcome of patients with metastatic and advanced-stage esophageal squamous cell carcinoma (ESCC). While cell behavior can be modulated by long non-coding RNAs (lncRNAs), the roles of lncRNAs within extracellular vesicles (exosomes) are largely unknown. Therefore, we investigated the involvement and regulatory functions of potential lncRNAs enclosed in exosomes during formation of chemoresistance in human ESCC. METHODS: Gefitinib-resistant cell lines were established by continuously grafting TE1 and KYSE-450 cells into gefitinib-containing culture medium. LncRNA microarray assay followed by RT-qPCR were used to verify the differential expression of lncRNA Prostate Androgen-Regulated Transcript 1 (PART1) between gefitinib resistant and parental cell lines. RNA fluorescence in situ hybridization (FISH) was used to investigate whether extracellular PART1 could be incorporated into exosomes and transmitted to recipient cells. Subsequently, a series of in vitro assays and a xenograft tumor model were used to observe the functions of lncRNA PART1 in ESCC cells. A signal transduction reporter array, bioinformatics analysis, western blotting, and immunofluorescence were carried out to verify the regulation of PART1 and its downstream Bcl-2 signaling pathway. RESULTS: lncRNA PART1 was upregulated in gefitinib-resistant cells when compared to parental ESCC cells. It was found that STAT1 can bind to the promoter region of lncRNA PART1, resulting in its activation. Knockdown of lncRNA PART1 potently promoted the gefitinib-induced cell death, while elevated PART1 promoted gefitinib resistance by competitively binding to miR-129 to facilitate Bcl-2 expression in ESCC cells. In addition, extracellular PART1 could be incorporated into exosomes and transmitted to sensitive cells, thus disseminating gefitinib resistance. Clinically, high levels of serum lncRNA PART1 in exosome were associated with poor response to gefitinib treatment in ESCC patients. CONCLUSIONS: LncRNA PART1 promotes gefitinib resistance by regulating miR-129/Bcl-2 pathway, and may serve as a therapeutic target for ESCC patients.


Subject(s)
Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Exosomes , Gefitinib/pharmacology , RNA, Untranslated/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Untranslated/biosynthesis , Signal Transduction , Transfection
7.
Oxid Med Cell Longev ; 2017: 7180943, 2017.
Article in English | MEDLINE | ID: mdl-29057033

ABSTRACT

Methylglyoxal (MGO), an active metabolite of glucose, has been reported to induce vascular cell apoptosis in diabetic complication. Polydatin (PD), a small natural compound from Polygonum cuspidatum, has a number of biological functions, such as antioxidative, anti-inflammatory, and nephroprotective properties. However, the protective effects of PD on MGO-induced apoptosis in endothelial cells remain to be elucidated. In this study, human umbilical vein endothelial cells (HUVECs) were used to explore the effects of PD on MGO-induced cell apoptosis and the possible mechanism involved. HUVECs were pretreated with PD for 2 h, followed by stimulation with MGO. Then cell apoptosis, reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP) impairment, mitochondrial morphology alterations, and Akt phosphorylation were assessed. The results demonstrated that PD significantly prevented MGO-induced HUVEC apoptosis. PD pretreatment also significantly inhibited MGO-induced ROS production, MMP impairment, mitochondrial morphology changes, and Akt dephosphorylation. These results and the experiments involving N-acetyl cysteine (antioxidant), Cyclosporin A (mitochondrial protector), and LY294002 (Akt inhibitor) suggest that PD prevents MGO-induced HUVEC apoptosis, at least in part, through inhibiting oxidative stress, maintaining mitochondrial function, and activating Akt pathway. All of these data indicate the potential application of PD for the treatment of diabetic vascular complication.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/therapeutic use , Endothelial Cells/metabolism , Glucosides/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Mitochondria/metabolism , Stilbenes/therapeutic use , Drugs, Chinese Herbal/pharmacology , Glucosides/pharmacology , Humans , Oxidative Stress , Stilbenes/pharmacology
8.
J Am Heart Assoc ; 6(7)2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28673898

ABSTRACT

BACKGROUND: Coagulation factor V (FV) plays a key role in hemostasis, is present in plasma and platelets, and has both pro- and anticoagulant properties; however, the contribution of platelet-derived FV to arterial thrombosis remains undetermined. METHODS AND RESULTS: Using transgenic mice with various levels of FV gene expression that was restricted to the plasma or platelets, the roles of platelet FV were evaluated in the regulation of arterial thrombosis and platelet activation. Mice with higher levels of platelet FV exhibited faster thrombotic occlusion of the carotid artery after injury compared with mice with lower platelet FV levels. Infusion of platelets with higher levels of FV into transgenic mice with undetectable levels of platelet FV reduced the time to carotid artery occlusion. In contrast, infusion of purified recombinant plasma FV into mice with undetectable platelet FV levels failed to reduce the carotid occlusion times following injury. Evaluation of isolated platelets revealed that platelet-derived FV was critical for the regulation of platelet activation. These effects were associated with an increased level of expression of P-selectin and increased cGMP in platelets. CONCLUSIONS: We established that platelet-derived FV is a critical mediator of arterial thrombosis that involves platelet activation.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Carotid Artery Diseases/blood , Factor V/metabolism , Platelet Activation , Thrombosis/blood , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Blood Platelets/drug effects , Carotid Artery Diseases/genetics , Cyclic GMP/blood , Disease Models, Animal , Factor V/administration & dosage , Factor V/genetics , Genetic Predisposition to Disease , Infusions, Intravenous , Male , Mice, Knockout , Phenotype , Platelet Activation/drug effects , Platelet Activation/genetics , Recombinant Proteins/administration & dosage , Selenoprotein P/blood , Thrombosis/genetics , Time Factors , Transforming Growth Factor beta
9.
Sci Rep ; 7: 43427, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28230206

ABSTRACT

Metformin, an anti-diabetic drug commonly used for type 2 diabetes therapy, is associated with anti-angiogenic effects in conditions beyond diabetes. miR-21 has been reported to be involved in the process of angiogenesis. However, the precise regulatory mechanisms by which the metformin-induced endothelial suppression and its effects on miR-21-dependent pathways are still unclear. Bioinformatic analysis and identification of miR-21 and its targets and their effects on metformin-induced antiangiogenic activity were assessed using luciferase assays, quantitative real-time PCR, western blots, scratch assays, CCK-8 assays and tubule formation assays. In this study, miR-21 was strikingly downregulated by metformin in a time- and dose-dependent manner. miR-21 directly targeted the 3'-UTR of PTEN and SMAD7, and negatively regulated their expression. Overexpression of miR-21 abrogated the metformin-mediated inhibition of endothelial cells proliferation, migration, tubule formation and the TGF-ß-induced AKT, SMAD- and ERK-dependent phosphorylations, and conversely, down-regulation of miR-21 aggravated metformin's action and revealed significant promotion effects. Our study broadens our understanding of the regulatory mechanism of miR-21 mediating metformin-induced anti-angiogenic effects, providing important implications regarding the design of novel miRNA-based therapeutic strategies against angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Smad7 Protein/genetics , 3' Untranslated Regions , Cell Movement/drug effects , Cell Proliferation/drug effects , Computational Biology , Gene Expression Regulation , Genes, Reporter , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Metformin , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Smad7 Protein/antagonists & inhibitors , Smad7 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
10.
J Vasc Res ; 53(5-6): 245-254, 2016.
Article in English | MEDLINE | ID: mdl-27894114

ABSTRACT

BACKGROUND: The flowers of Carthamus tinctorius L. are widely used in traditional Chinese medicine to treat cerebrovascular and cardiovascular diseases. Hydroxysafflor yellow A (HSYA), the main constituent of C. tinctorius L. flowers, is known for its multiple biological activities. The present study investigated the effects of HSYA on angiogenesis in vitro and in a mouse hindlimb ischemia model. METHODS: Using human umbilical vein endothelial cells (HUVEC) in vitro and a mouse hindlimb ischemia model in vivo, the angiogenic role of HSYA was evaluated. RESULTS: HSYA significantly increased the capillary-like tube formation and migration of HUVEC. HSYA not only induced a rise in the expression of angiopoietin 1 and Tie-2 but it also increased phosphorylation of Tie-2, Akt, and extracellular signal-regulated kinase 1/2. Furthermore, an anti-Tie-2 neutralizing antibody significantly inhibited HSYA-induced HUVEC tube formation and migration. In vivo, the recovery of perfusion of ischemic hindlimb tissue after femoral artery interruption was significantly increased in HSYA-treated mice compared to vehicle controls. Consistent with these results, the arteriole and capillary densities in ischemic gastrocnemius muscles were significantly increased in HSYA-treated mice. CONCLUSIONS: These results indicate the potential utility of HSYA for the treatment of ischemic diseases.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Angiopoietin-1/metabolism , Chalcone/analogs & derivatives , Human Umbilical Vein Endothelial Cells/drug effects , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Quinones/pharmacology , Receptor, TIE-2/metabolism , Signal Transduction/drug effects , Animals , Blood Flow Velocity , Cell Movement/drug effects , Cells, Cultured , Chalcone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Regional Blood Flow , Time Factors
11.
Sci Rep ; 6: 36687, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819307

ABSTRACT

Type 2 diabetic mellitus (DM2) is associated with accelerated thrombotic complications and is characterized by high levels of plasminogen activator inhibitor-1 (PAI-1). Recent studies show that human platelets have high levels of miR-30c and synthesize considerable active PAI-1. The underlying mechanism of how PAI-1 expression is upregulated in DM2 is poorly understood. We now report that hyperglycaemia-induced repression of miR-30c increases PAI-1 expression and thrombus formation in DM2. Bioinformatic analysis and identification of miRNA targets were assessed using luciferase assays, quantitative real-time PCR and western blots in vitro and in vivo. The changes in miR-30c and PAI-1 levels were identified in platelets from healthy and diabetic individuals. We found that miR-30c directly targeted the 3' UTR of PAI-1 and negatively regulated its expression. miR-30c was negatively correlated with glucose and HbA1c levels in DM2. In HFD-fed diabetic mice, increasing miR-30c expression by lenti-miR-30c significantly decreased the PAI-1 expression and prolonged the time to occlusion in an arterial thrombosis model. Platelet depletion/reinfusion experiments generating mice with selective ablation of PAI-1 demonstrate a major contribution by platelet-derived PAI-1 in the treatment of lenti-miR-30c to thrombus formation. These results provide important implications regarding the regulation of fibrinolysis by platelet miRNA under diabetic mellitus.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 2/metabolism , Hyperglycemia/metabolism , MicroRNAs/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Animals , Cells, Cultured , Humans , Male , Mice, Inbred C57BL
12.
J Transl Med ; 14(1): 306, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27784306

ABSTRACT

BACKGROUND: Angiopoietin-2 (Ang-2), a ligand of the Tie-2 receptor, plays an important role in maintaining endothelial cells and in destabilizing blood vessels. Collateral artery growth (arteriogenesis) is a key adaptive response to arterial occlusion. It is unknown whether the destabilization of blood vessels by Ang-2 can affect arteriogenesis and modulate mononuclear cell function. This study aimed to investigate the effects of Ang-2 on collateral artery growth. METHODS: Hindlimb ischaemia model was produced in C57BL/6 mice by femoral artery ligation. Blood flow perfusion was measured using a laser Doppler perfusion imager quantitative RT-PCR analysis was applied to identify the level of angiogenic factors. RESULTS: After the induction of hindlimb ischaemia, blood flow recovery was impaired in mice treated with recombinant Ang-2 protein; this was accompanied by a reduction of peri-collateral macrophage infiltration. In addition, quantitative RT-PCR analysis revealed that Ang-2 treatment decreased monocyte chemotactic protein-1 (MCP-1), platelet-derived growth factor-BB (PDGF-BB) mRNA levels in ischaemic adductor muscles. Ang-2 can lead to macrophage M1/M2 polarization shift inhibition in the ischaemic muscles. Furthermore, Ang-2 reduced the in vitro inflammatory response in macrophages and vascular cells involved in arteriogenesis. CONCLUSIONS: Our results demonstrate that Ang-2 is essential for efficient arteriogenesis, which controls macrophage infiltration.


Subject(s)
Angiopoietin-2/therapeutic use , Arteries/growth & development , Collateral Circulation , Hindlimb/blood supply , Hindlimb/pathology , Ischemia/drug therapy , Macrophages/pathology , Angiopoietin-2/pharmacology , Animals , Arteries/drug effects , Arteries/pathology , Cell Movement/drug effects , Collateral Circulation/drug effects , Gene Expression Regulation/drug effects , Humans , Inflammation/pathology , Ischemia/genetics , Ischemia/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Perfusion , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
13.
Microvasc Res ; 105: 70-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26808210

ABSTRACT

Diabetes mellitus (DM) leads to the development of microvascular diseases and is associated with impaired angiogenesis. The presence of vascular endothelial growth factor (VEGF) can block PDGF-BB dependent regulation of neovascularization and vessel normalization. We tested the hypothesis that the inhibition of VEGF improves blood flow in a mouse hindlimb ischemia model produced by femoral artery ligation. In this study, we examined the effect of bevacizumab, a humanized monoclonal antibody against VEGF-A, on blood perfusion and angiogenesis after hindlimb ischemia. We showed that bevacizumab induces functional blood flow in high fat chow (HFC)-fed diabetic mice. Treatment with bevacizumab increased the expression of platelet derived growth factor-BB (PDGF-BB) in ischemic muscle, and led to vascular normalization. It also blocked vascular leakage by improving the recruitment of pericytes associated with nascent blood vessels, but it did not affect capillary formation. Furthermore, treatment with an anti-PDGF drug significantly inhibited blood flow perfusion in diabetic mice treated with bevacizumab. These results indicate that bevacizumab improves blood flow recovery through the induction of PDGF-BB in a diabetic mouse hindlimb ischemia model, and that vessel normalization may represent a useful strategy for the prevention and treatment of diabetic peripheral arterial disease.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Diabetic Angiopathies/drug therapy , Diet, High-Fat , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Neovascularization, Pathologic , Peripheral Arterial Disease/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Remodeling/drug effects , Animals , Becaplermin , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Disease Models, Animal , Hindlimb , Ischemia/etiology , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Peripheral Arterial Disease/etiology , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/physiopathology , Proto-Oncogene Proteins c-sis/metabolism , Regional Blood Flow , Signal Transduction/drug effects , Time Factors , Vascular Endothelial Growth Factor A/metabolism
14.
Mol Cancer ; 14: 140, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26215730

ABSTRACT

BACKGROUND: An increased incidence of venous thromboembolism (VTE) is associated with anti-vascular endothelial growth factor (VEGF) treatment in cancer. However, the mechanism underlying this effect remains elusive. In this study, we examined the effect of bevacizumab, a humanized monoclonal antibody against VEGF-A, on VTE in a murine xenograft A549 cell tumor model. METHODS: Inferior vena cava stenosis model and FeCl3-induced saphenous vein thrombosis model were performed in a mouse xenograft models of human lung adenocarcinoma. RESULTS: We found that treatment with bevacizumab significantly increased the thrombotic response to inferior vena cava obstruction and femoral vein injury. Plasminogen activator inhibitor (PAI-1) expression in tumors, plasma, and thrombi was significantly increased by bevacizumab. However, bevacizumab did not enhance VTE in PAI-1-deficient mice, suggesting that PAI-1 is a major mediator of bevacizumab's prothrombotic effect. VEGF inhibited expression of PAI-1 by A549 cells, and this effect was neutralized by bevacizumab, suggesting that bevacizumab increases PAI-1 expression in vivo by blocking the inhibitory effect of VEGF on PAI-1 expression by tumor cells. Pharmacological inhibition of PAI-1 with PAI-039 blocked bevacizumab-induced venous thrombosis. CONCLUSION: Collectively, these findings indicate that PAI-1 plays a role in VTE associated with antiangiogenic therapy and the inhibition of PAI-1 shows efficacy as a therapeutic strategy for the prevention of bevacizumab-associated VTE.


Subject(s)
Adenocarcinoma/pathology , Bevacizumab/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Plasminogen Activator Inhibitor 1/biosynthesis , Venous Thromboembolism/pathology , Adenocarcinoma/complications , Adenocarcinoma/drug therapy , Adenocarcinoma of Lung , Animals , Bevacizumab/administration & dosage , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Mice , Plasminogen Activator Inhibitor 1/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Venous Thromboembolism/chemically induced , Venous Thromboembolism/etiology , Xenograft Model Antitumor Assays
15.
Acta Diabetol ; 52(5): 943-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25820527

ABSTRACT

AIMS: MicroRNA-103 (miR-103) plays a critical role in regulating glucose homeostasis in type 2 diabetes (DM2). Recent data suggest that secreted frizzled-related protein 4 (SFRP4) serves as a potential risk biomarker for prediabetic mellitus (pre-DM) and that platelets are enriched for miR-103. The objective of this study was to test the hypothesis that platelet-derived miR-103b (miR-103-as), which regulates SFRP4, might be a novel biomarker for the early diagnosis of DM2. METHODS: We evaluated platelet miR-103b expression in healthy subjects (n = 46), pre-DM subjects (n = 48), non-complicated diabetic subjects (n = 43) and diabetes mellitus type 2-coronary heart disease subjects (n = 36), respectively, and analyzed the relationship of these levels with its target gene SFRP4. RESULTS: In qRT-PCR assays, miR-103b were significantly down-regulated, and conversely, the expression of the SFRP4 gene was up-regulated in pooled leukocyte-depleted platelets and individual subjects with pre-DM. Additionally, patients who had undergone antiplatelet treatment were characterized by decreased gene expression of SFRP4 and increased levels of platelet-derived miR-103b. miR-103b modulated reporter gene expression through SFRP4 mRNA 3'-UTR seed sequence and negatively regulated its expression. Furthermore, SFRP4 mRNA and protein levels were down-regulated by a miR-103b mimic but were up-regulated by a miR-103b inhibitor. CONCLUSIONS: The results suggest that platelet-derived miR-103b could negatively regulate the expression of SFRP4 mRNA/protein in pre-DM2, indicating that miR-103b could be a novel biomarker for the early diagnosis of DM2.


Subject(s)
Blood Platelets/chemistry , Diabetes Mellitus, Type 2/diagnosis , MicroRNAs/blood , 3' Untranslated Regions/genetics , Adult , Aged , Biomarkers/blood , Cell Line , Coronary Disease/complications , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Early Diagnosis , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics
16.
Int J Clin Exp Med ; 8(11): 20463-71, 2015.
Article in English | MEDLINE | ID: mdl-26884962

ABSTRACT

This study is to investigate the effect and underlying mechanism of Zinc (Zn) on hepatic stellate cell collagen synthesis. The proliferation and collagen synthesis ability of LX-2 cells were detected after adding Zn. The collagen synthesis related proteins of MMP-13 and TIMP1 along with TGF-ß signaling pathway related proteins were detected by Western blot. The role of TGF-ß signaling pathway in collagen synthesis inhibition was identified by TGF-ß RI siRNA silencing. Compared with control group, LX-2 cell proliferation ability was significantly inhibited at all Zn concentrations (50 µM, 100 µM and 200 µM). Zn at 50 µM did not affect the protein content of αSMA and type I collagen while 100 µM and 200 µM Zn could significantly inhibit αSMA expression. Compared with control group, gene expression and protein content of MMP-13 in 200 µM Zn group was significantly increased while no difference in gene expression and protein content of TIMP1 was found. TGF-ß RI content in 200 µM Zn group was significantly decreased and the protein content of TGF-ß RII was not affected. MMP-13 expression was significantly increased after TGF-ß RI siRNA silencing. Further results showed that in LX-2 cells those TGF-ß RI expression was inhibited, LX-2 cell proliferation ability and the expression of synthesis collagen related proteins of αSMA and type I collagen were greatly decreased. Zn could significantly inhibit the expression of αSMA and type I collagen by inhibiting TGF-ß RI expression and promoting MMP-13 expression.

17.
Int J Clin Exp Med ; 8(11): 20603-9, 2015.
Article in English | MEDLINE | ID: mdl-26884979

ABSTRACT

The present study is to investigate the molecular mechanism by which Zinc (Zn) deficiency induces apoptosis in hepatic stellate cells. LX-2 cells were incubated with N,N,N',N'-tetrakis(2-pyridylmethyl)ethane-1,2-diamine (TPEN; 5 µM, 10 µM and 25 µM) for 24 h. MTT assay was used to test the proliferation ability of LX-2 cells. Flow cytometry was performed to detect cell apoptosis. Western blotting assay was employed to determine the expression of metallothionein (MT). Atomic absorption spectroscopy was performed to measure intracellular reactive oxygen species content. To test the activity of mitochondria, respiratory control rate was tested. To investigate the activation of apoptotic signaling pathway, cytochrome C oxidase activity was determined. TPEN effectively decreased the content of Zn in LX-2 cells. Zn deficiency led to the inhibition of proliferation and enhancement of apoptosis of LX-2 cells. Zn deficiency induced the inhibition of MT expression in LX-2 cells. Inhibition of MT expression induced by Zn deficiency resulted in enhanced reactive oxygen species content, impaired mitochondrial function and inhibition of cytochrome C oxidase activity. Intracellular MT content in LX-2 cells is reduced by Zn deficiency. Reduction in MT expression further increases intracellular ROS content, enhances oxidative stress, inhibits cytochrome C oxidase activity, impairs mitochondrial function, and finally leads to cell apoptosis.

18.
Am J Transl Res ; 7(11): 2212-22, 2015.
Article in English | MEDLINE | ID: mdl-26807169

ABSTRACT

MicroRNAs (miRNAs) play important roles in cancer progression including gastric cancer. miR-485-5p is reported as a potential suppressor in breast cancer, but its expression, cellular function and clinic features in gastric cancer is not known. In our study, we found that miR-485-5p expression was down-regulated in gastric cancer cell lines. miR-485-5p could inhibit gastric cancer cell growth in vitro and in vivo. We also found that miR-485-5p suppressed gastric cancer cell metastasis and sphere formation. It was confirmed flotillin-1 (Flot1) as a direct target of miR-485-5p, and up-regulation of miR-485-5p could decrease expression of Flot1 in gastric cancer cells. Further investigation showed that ectopic expression of Flot1 partially reversed the inhibition effect of enforced miR-485-5p expression on the malignant phenotypes of gastric cancer cells. The low expression of miR-485-5p in gastric cancer tissues was related to advanced clinical features and poorer prognosis. Our study suggested that miR-485-5p could be a potential prognostic marker and functions as a tumor suppressor in human gastric cancer by post-transcriptionally targeting Flot1.

19.
Arterioscler Thromb Vasc Biol ; 35(1): 111-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25378411

ABSTRACT

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1) regulates angiogenesis via effects on extracellular matrix proteolysis and cell adhesion. However, no previous study has implicated PAI-1 in controlling vascular endothelial growth factor (VEGF) signaling. We tested the hypothesis that PAI-1 downregulates VEGF receptor-2 (VEGFR-2) activation by inhibiting a vitronectin-dependent cooperative binding interaction between VEGFR-2 and αVß3. APPROACH AND RESULTS: We studied effects of PAI-1 on VEGF signaling in human umbilical vein endothelial cells. PAI-1 inhibited VEGF-induced phosphorylation of VEGFR-2 in human umbilical vein endothelial cells grown on vitronectin, but not on fibronectin or collagen. PAI-1 inhibited the binding of VEGFR-2 to ß3 integrin, VEGFR-2 endocytosis, and intracellular signaling pathways downstream of VEGFR-2. The anti-VEGF effect of PAI-1 was mediated by 2 distinct pathways, one requiring binding to vitronectin and another requiring binding to very low-density lipoprotein receptor. PAI-1 inhibited VEGF-induced angiogenesis in vitro and in vivo, and pharmacological inhibition of PAI-1 promoted collateral arteriole development and recovery of hindlimb perfusion after femoral artery interruption. CONCLUSIONS: PAI-1 inhibits activation of VEGFR-2 by VEGF by disrupting a vitronectin-dependent proangiogenic binding interaction involving αVß3 and VEGFR-2. These results broaden our understanding of the roles of PAI-1, vitronectin, and endocytic receptors in regulating VEGFR-2 activation and suggest novel therapeutic strategies for regulating VEGF signaling.


Subject(s)
Endothelial Cells/metabolism , Integrin alphaVbeta3/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Plasminogen Activator Inhibitor 1/metabolism , Receptor Cross-Talk , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Disease Models, Animal , Endocytosis , Endothelial Cells/drug effects , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoleacetic Acids/administration & dosage , Ischemia/metabolism , Ischemia/physiopathology , Ischemia/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neovascularization, Physiologic/drug effects , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , RNA Interference , Receptor Cross-Talk/drug effects , Receptors, LDL/metabolism , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Time Factors , Transfection , Vitronectin/deficiency , Vitronectin/genetics
20.
Am J Physiol Regul Integr Comp Physiol ; 307(7): R901-7, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25275013

ABSTRACT

It is known that Toll-like receptor (TLR)-4 plays an important role in myocardial infarction and atherothrombosis. The role of TLR-4 in arterial thrombosis is undefined. Both TLR-4-deficient (TLR-4(-/-)) and wild-type (WT) mice were subjected to FeCl3 carotid artery injury, and the time required to form an occlusive thrombus was measured. The mean time to occlusion in TLR-4(-/-) mice was significantly greater than that in WT mice after injury (303 ± 32 vs. 165 ± 34 s, P < 0.05). Furthermore, when we used a WT or TLR-4(-/-)-derived platelet reinfusion in a platelet depletion/reinfusion procedure, there was no significant change in the occlusion time and tissue factor (TF) activity in injured arteries between WT mice and platelet-depleted WT mice. Similarly, no significant difference was observed between TLR-4(-/-) mice and platelet-depleted TLR-4(-/-) mice for the WT or TLR-4(-/-)-derived platelet reinfusion. However, TF expression and activity were significantly reduced in the vascular wall of TLR-4(-/-) mice compared with WT mice. In vivo, lipopolysaccharide accelerated the occlusion time in WT mice but not TLR-4(-/-) mice. In vitro, LPS-induced TF activity was reduced in endothelial cells of TLR-4(-/-) mice relative to WT mice. The data demonstrate that TLR-4 contributes to arterial thrombosis formation in vivo and causes increased TF expression and activity in vitro. The results further suggest that the stimulation is mainly derived by endothelial cells but is not due to platelet-derived TLR-4.


Subject(s)
Endothelial Cells/metabolism , Thromboplastin/metabolism , Thrombosis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Blood Platelets/metabolism , Carotid Artery Injuries/chemically induced , Disease Models, Animal , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Thromboplastin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...