Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Eur J Pharmacol ; 972: 176543, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38582274

ABSTRACT

Cyclosporin A, an immunosuppressive agent, is extensively utilized for the prevention of transplant rejection and treat autoimmune disease in the clinic, despite its association with a high risk of hypertension development among patients. Resveratrol is a kind of non-flavonoid phenolic compound that widely exists in many plants. The aim of the present study was to investigate the mechanism by which resveratrol ameliorates cyclosporin A-induced hypertension. The arterial rings of the mesentery were incubated with cyclosporin A and resveratrol in vitro. Rats were administered cyclosporin A and/or resveratrol for 3 weeks in vivo. Blood pressure was measured via the tail arteries. Vasoconstriction curves were recorded using a sensitive myograph. The protein expression was evaluated through Western blotting. This study demonstrated that resveratrol mitigated the cyclosporin A-induced increase in blood pressure in rats. Furthermore, resveratrol markedly inhibited the cyclosporin A-induced upregulation of thromboxane A2 receptor-mediated vasoconstriction in the rat mesenteric artery both in vitro and in vivo. Moreover, resveratrol activated AMPK/SIRT1 and inhibited the MAPK/NF-κB signaling pathway. In conclusion, resveratrol restored the cyclosporin A-induced upregulation of the thromboxane A2 receptor and hypertension via the AMPK/SIRT1 and MAPK/NF-κB pathways in rats.


Subject(s)
AMP-Activated Protein Kinases , Cyclosporine , Hypertension , Mesenteric Arteries , NF-kappa B , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1 , Up-Regulation , Animals , Resveratrol/pharmacology , Cyclosporine/pharmacology , Sirtuin 1/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Male , NF-kappa B/metabolism , Up-Regulation/drug effects , Rats , AMP-Activated Protein Kinases/metabolism , Vasoconstriction/drug effects , Blood Pressure/drug effects , Signal Transduction/drug effects , Mitogen-Activated Protein Kinases/metabolism
2.
Chem Biol Interact ; 368: 110203, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36183783

ABSTRACT

Smoking is a well-known risk factor for cardiovascular diseases. However, the mechanisms behind how smoking leads to vasospasm are not fully elucidated. Endothelin receptors are involved in the pathogenesis of cardiovascular diseases. This study examined whether cigarette smoke could induce up-regulation of vascular endothelin receptors through AMPK-SIRT1 and MAPK pathways. The results show that DMSO-soluble smoking particles (DSP) up-regulated the protein expressions of endothelin receptors and the contractile responses. Furthermore, the inhibition of MAPK or activation of AMPK-SIRT1 markedly attenuated DSP-enhanced vasoconstriction and the protein expression of endothelin receptors. The in vivo study showed that cigarette smoke increased the blood pressure of the rats and down-regulated p-AMPKα, SIRT1, and up-regulated endothelin receptors, p-ERK1/2, and p-P38 protein expressions. However, treatment with resveratrol attenuated vasoconstriction, endothelin receptor proteins expression, and blood pressure in vivo. In conclusion, this suggests that cigarette smoke up-regulates the vascular endothelin receptors through AMPK-SIRT1 and MAPK pathways.


Subject(s)
Cardiovascular Diseases , Receptors, Endothelin , Rats , Animals , Receptors, Endothelin/metabolism , Sirtuin 1 , Dimethyl Sulfoxide , AMP-Activated Protein Kinases , Rats, Sprague-Dawley , Nicotiana/metabolism , Smoking
3.
Eur J Pharmacol ; 926: 175034, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35588871

ABSTRACT

Cyclosporin A (CsA) is an immunosuppressant used in transplantation patients and inflammatory diseases. CsA-induced local vasoconstriction can lead to serious side effects including nephrotoxicity and hypertension. However, the underlying mechanisms are not fully understood. Mesenteric artery rings of rats were cultured with CsA and specific inhibitors for mitogen-activating protein kinases (MAPK) and nuclear factor-κB (NF-κB) signaling pathways. A sensitive myograph recorded thromboxane (TP) receptor-mediated vasoconstriction. Protein levels of key signaling molecules were assessed by Western blotting. The results show that CsA up-regulated the TP receptor expression with the enhanced vasoconstriction in a dose- and time-dependent manner. Furthermore, the blockage of MAPKs or NF-κB activation markedly attenuated CsA-enhanced vasoconstriction and the TP receptor protein expression. Rats subcutaneously injected with CsA for three weeks showed increased blood pressure in vivo and increased contractile responses to a TP agonist ex vivo. CsA also enhanced TP receptor, as well as p-ERK1/2, p-p38, p- IκBα, p-NF-κB P65 protein levels and decreased IκBα protein expression, demonstrating that CsA induced TP receptor enhanced-vasoconstriction via activation of MAPK and NF-κB pathways. In conclusion, CsA up-regulated the expression of TP receptors via activation of MAPK and NF-κB pathways. The results may provide novel options for prevention of CsA-associated hypertension.


Subject(s)
Cyclosporine , Hypertension , NF-kappa B , Receptors, Thromboxane A2, Prostaglandin H2 , Animals , Cyclosporine/pharmacology , Humans , Mesenteric Arteries/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mitogens , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Rats , Receptors, Thromboxane , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Thromboxanes , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Microvasc Res ; 139: 104260, 2022 01.
Article in English | MEDLINE | ID: mdl-34624308

ABSTRACT

OBJECTIVES: To explore whether minimally modified low-density lipoprotein (mmLDL) upregulates mesenteric arterial 5-hydroxytryptamine 1B (5-HT1B) receptor expression by activating the JAK2/STAT3 signaling pathway. METHODS: Mice were randomly divided into the following groups: the normal saline (NS), LDL, mmLDL, mmLDL+galiellactone (GL, a JAK2/STAT3 pathway inhibitor), and mmLDL+DMSO groups. The dose-response curve of mesenteric arterial ring constriction after administration of 5-carboxamidotryptamine (5-CT), an agonist of 5-HT1B, was recorded with a microvascular tensiometer. JAK2, p-JAK2, STAT3, p-STAT3, and 5-HT1B receptor protein expression levels were determined by Western blotting. 5-HT1B receptor mRNA levels were measured by RT-PCR. 5-HT1B receptor protein expression was determined by immunofluorescence. RESULTS: Injection of mmLDL into the tail vein significantly increased the contractile dose-response curve after 5-CT stimulation, as the Emax was 82.15 ±â€¯6.15% in the NS group and 171.88 ±â€¯5.78% in the mmLDL group (P < 0.01); significantly elevated 5-HT1B receptor mRNA and protein expression levels; and significantly increased p-JAK2 and p-STAT3 protein expression levels. After intraperitoneal injection of GL, the vasoconstrictive response was significantly reduced compared with that in the mmLDL group, as the Emax was decreased to 97.14 ±â€¯1.20% (P < 0.01); 5-HT1B receptor mRNA and protein expression levels were significantly reduced; STAT3 phosphorylation and p-JAK2 and p-STAT3 protein expression were not significantly changed; and 5-HT1B receptor expression was altered via inhibition of p-STAT3 binding to DNA, which suppressed transcription. CONCLUSIONS: mmLDL can upregulate 5-HT1B receptor expression in mouse mesenteric arteries by activating the JAK2/STAT3 signaling pathway.


Subject(s)
Janus Kinase 2/metabolism , Lipoproteins, LDL/pharmacology , Mesenteric Arteries/drug effects , Receptor, Serotonin, 5-HT1B/metabolism , STAT3 Transcription Factor/metabolism , Vasoconstriction/drug effects , Animals , Enzyme Activation , Female , Male , Mesenteric Arteries/enzymology , Mice , Phosphorylation , Receptor, Serotonin, 5-HT1B/genetics , Signal Transduction , Up-Regulation
5.
Pathol Res Pract ; 216(9): 153083, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32825951

ABSTRACT

Chronic exposure to cigarette smoke is the major risk factor for the development of pulmonary hypertension (PH) with chronic lung disease (i.e. PH group III). The pathogenesis of smoke-associated PH group III in chronic obstructive pulmonary disease (COPD) involves cigarette smoke exposure-induced damage to lung tissue and dysfunction of pulmonary system with increased synthesis and release of endothelin-1 (ET-1), hypoxia, inflammation, pulmonary vascular remodeling. Many studies have demonstrated that cigarette smoke exposure induces activation of mitogen-activated protein kinase (MAPK) signal pathway that leads to up-regulation of ET-1 and its receptors with the receptor-mediated enhanced contraction, proliferation of pulmonary vascular smooth muscle cells, pulmonary vascular remodeling, elevated pulmonary arterial pressure and finally PH group III. This mini-review article aims to summarize the current state of understanding on the roles of cigarette smoke-induced up-regulation of ET-1 and its receptors in the development of PH group III. Understanding the underlying molecular mechanisms that cigarette smoke exposure leads to PH group III may provide a novel strategy for the treatment.


Subject(s)
Hypertension, Pulmonary/pathology , Lung Diseases/pathology , Lung/pathology , Muscle, Smooth, Vascular/pathology , Animals , Endothelins/metabolism , Humans , Hypertension, Pulmonary/metabolism , Lung/metabolism , Lung Diseases/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction/physiology
6.
Int J Nanomedicine ; 15: 5405-5416, 2020.
Article in English | MEDLINE | ID: mdl-32801696

ABSTRACT

PURPOSE: Although the effective and safe medical defoamers, dimethicone (DM) and simethicone (SM) are widely used in electronic gastroscope examination (EGE), their preparations are presented in the form of suspensions or emulsions, these are untransparent or milk-like in appearance and can easily cause misdiagnosis as a result of an unclear field of vision if the doctor does not master the amount of defoamer or operates incorrectly. At the same time, it is also difficult to wash out the camera and pipeline, due to the large oil droplets of preparations. The purpose of this study was to develop a new clear and transparent oil in water (O/W) DM nanoemulsions (DMNs) and observe the effect of application in EGE. METHODS: The oil phase was chosen for its antifoaming activity and viscosity. The emulsifier and co-emulsifier were selected according to the solubility of the oil phase in them. The water titration method was used to make the pseudoternary phase diagrams of nanoemulsions and optimize the prescription composition. DM-in-water nanoemulsion was prepared by the low energy method and evaluated for appearance, antifoaming ability, droplet size, and stability. The effect of DMNs utilized in EGEs was also observed. RESULTS: The optimal formulation of DMNs contained CRH-40 as an emulsifier, PEG-400 as a co-emulsifier, DM as oil phase with the viscosity of 10 mPa.s, and their proportion was 4.5:4.5:1, respectively. DMNs obtained the average particle size of 67.98 nm with the polydispersity index (PDI) of 0.332, and 57.14% defoaming rate. The result of using an EGE showed that DMNs were superior in comparison to the emulsions with regard to the defoaming effect, visual clarity, and easy cleanup. CONCLUSION: DMNs were found to provide excellent visual clarity to its other preparations. The novel DMNs is a promising substitute for DM emulsions or suspensions in EGEs.


Subject(s)
Antifoaming Agents/chemistry , Dimethylpolysiloxanes/chemistry , Emulsions/chemistry , Gastroscopy/methods , Antifoaming Agents/adverse effects , Antifoaming Agents/therapeutic use , Castor Oil/chemistry , Dimethylpolysiloxanes/adverse effects , Dimethylpolysiloxanes/therapeutic use , Emulsifying Agents/chemistry , Female , Gastroscopy/adverse effects , Humans , Male , Nanostructures/chemistry , Particle Size , Polyethylene Glycols/chemistry , Solubility , Viscosity
7.
Adv Pharmacol Pharm Sci ; 2020: 5070436, 2020.
Article in English | MEDLINE | ID: mdl-32309807

ABSTRACT

Minimally modified low-density lipoprotein (mmLDL) is a risk factor for cardiovascular diseases. The current study explored the effect of mmLDL on the endothelin type A (ETA) receptor in mouse mesenteric arteries in vivo, as well as the role of autophagy in this process. mmLDL was injected via the caudal vein, and the Class III PI3K autophagic pathway inhibitor 3-methyladenine (3-MA) was injected intraperitoneally. The animals were divided into physiological saline (NS), mmLDL, and mmLDL + 3-MA groups. The dose-effect curve of endothelin-1- (ET-1-) induced mesenteric artery contraction was measured using myography, while ETA receptor mRNA expression was detected using real-time polymerase chain reactions, and the protein levels of the ETA receptor, class III PI3K, Beclin-1, LC3 II/I, p62, NF-κB, and p-NF-κB were observed using Western blot analysis. mmLDL significantly strengthened ET-1-induced contraction (the E max value increased from 184.87 ± 7.46% in the NS group to 319.91 ± 20.31% in the mmLDL group (P < 0.001), and the pEC50 value increased from 8.05 ± 0.05 to 9.11 ± 0.09 (P < 0.01). In addition to upregulating the protein levels of Class III PI3K, Beclin-1, and LC3 II/I and downregulating that of p62, mmLDL significantly increased the mRNA expression and protein level of the ETA receptor and increased the protein level of p-NF-κB. However, these effects were significantly inhibited by 3-MA. mmLDL activates autophagy via the Class III PI3K/Beclin-1 pathway and upregulates the ETA receptor via the downstream NF-κB pathway. Understanding the effect of mmLDL on the ETA receptor and the underlying mechanisms may provide a new idea for the prevention and treatment of cardiovascular diseases.

8.
Microvasc Res ; 127: 103923, 2020 01.
Article in English | MEDLINE | ID: mdl-31494123

ABSTRACT

Minimally modified low-density lipoprotein (mmLDL) is a risk factor for cardiovascular disease. This study was designed to investigate the effect of a Toll-like receptor 4 monoclonal antibody (TLR4 mAb) on mmLDL-induced endothelium-dependent vasodilation (EDV) impairment in mouse mesenteric arteries and to explore the underlying mechanism. Animals were divided into a normal control group, an mmLDL treatment group, and a TLR4 mAb intervention group. The serum concentrations of IL-1ß and TNF-α were detected using enzyme-linked immunosorbent assays (ELISAs). EDV function was measured using a microvascular tension tracing method. The protein levels and mRNA expression of IL-1ß and TNF-α in vascular tissue were detected using western blot analysis and reverse transcription polymerase chain reaction, respectively. TLR4 mAb improved mmLDL-induced EDV functional impairment in a dose-dependent manner. TLR4 mAb significantly upregulated KCa3.1 and KCa2.3 channel protein levels and downregulated TNF-α and IL-1ß expression. These effects were possibly associated with the competitive antagonism of TLR4 mAb on the TLR4 signaling pathway and the downstream NF-κB p65 and p38 MAPK pathways, which are activated by mmLDL. In conclusion, pretreatment with TLR4 mAb lessens mmLDL-induced EDV dysfunction and inhibits overexpression of inflammatory factors. Regulation of the TLR4 pathway, as well as its downstream NF-κB p65 and p38 MAPK pathways, may be an effective strategy for the prevention and treatment of cardiovascular diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Endothelium, Vascular/drug effects , Lipoproteins, LDL/pharmacology , Mesenteric Arteries/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Vasodilation/drug effects , Animals , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Interleukin-1beta/blood , Interleukin-1beta/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mesenteric Arteries/immunology , Mesenteric Arteries/metabolism , Mice, Inbred ICR , Phosphorylation , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Biol Pharm Bull ; 42(5): 703-711, 2019.
Article in English | MEDLINE | ID: mdl-31061312

ABSTRACT

Cigarette smoke is a well-known strong risk factor for inducing airway hyperreactivity (AHR), but the underlying molecular mechanisms are not fully understood. In the present study, mouse in-vivo and in-vitro models were used to study effects of dimethyl sulfoxide (DMSO)-extracted cigarette smoke particles (DSP) on the airway, and to explore the underlying molecular mechanisms that are involved in DSP-induced AHR. In mouse in-vivo model, DSP (0.75, 1.5 or 3 µL/mL) was administered intranasally daily for 7 d. At the end of this period, lung functions were measured with flexiVent™. The results showed that the mice exhibited AHR in a dose-dependent manner following methacholine inhalation in vivo. In mouse in-vitro organ culture model, exposure of mouse tracheal segments to DSP (0.1 µL/mL) with or without the following pharmacological inhibitors: specific c-Jun-N-terminal kinase (JNK) inhibitor SP600125 (10 µM) or the anti-inflammatory drug dexamethasone (1 µM). DSP-induced bradykinin receptor-mediated airway contraction with increased mRNA and protein expressions for bradykinin B1 and B2 receptors could be significantly reduced by SP600125 or dexamethasone. In conclusion, the present study demonstrates that DSP could induce AHR in vivo and in vitro. In addition to this, the upregulation of bradykinin receptors in airway is most likely one of the underlying molecular mechanisms involved.


Subject(s)
Bronchial Hyperreactivity/chemically induced , Nicotiana/adverse effects , Smoke/adverse effects , Animals , Anthracenes/pharmacology , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Dimethyl Sulfoxide/chemistry , JNK Mitogen-Activated Protein Kinases/physiology , Male , Mice, Inbred BALB C , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Solvents/chemistry , Trachea/drug effects , Trachea/physiology
10.
J Pharm Pharmacol ; 71(6): 937-944, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30663067

ABSTRACT

OBJECTIVES: The current study aimed to explore whether minimally modified low-density lipoprotein (mmLDL) via tail vein injection upregulates the ETB and α1 receptors in mouse mesenteric arteries by activating the PI3K/Akt pathway. METHODS: The contraction curves of the mesenteric arteries caused by sarafotoxin 6c (S6c, ETB receptor agonist) and phenylephrine (PE, α1 receptor agonist) were measured by a myograph system. Serum oxLDL was detected using enzyme-linked immunosorbent assays. The levels of the ETB receptor, the α1 receptor, PI3K, p-PI3K and p-Akt were detected using real-time polymerase chain reaction and Western blot analyses. KEY FINDINGS: Minimally modified low-density lipoprotein noticeably enhanced the contraction effect curves of S6c and PE, with significantly increased Emax values (P < 0.01), compared to those of the control group. This treatment significantly increased the mRNA expression and protein levels of the ETB and α1 receptors and the protein levels of p-PI3K and p-Akt in the vessel wall (P < 0.01). LY294002 inhibited the effect of mmLDL. CONCLUSIONS: An increase in mmLDL activated the PI3K/Akt pathway, which upregulated the expression of the ETB and α1 receptors and enhanced the ETB and α1- receptor-mediated contractile function.


Subject(s)
Lipoproteins, LDL/blood , Mesenteric Arteries/metabolism , Receptor, Endothelin B/genetics , Receptors, Adrenergic, alpha-1/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Male , Mesenteric Arteries/drug effects , Mice , Mice, Inbred ICR , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Endothelin B/agonists , Receptors, Adrenergic, alpha-1/drug effects , Up-Regulation , Vasoconstrictor Agents/pharmacology , Viper Venoms/pharmacology
11.
Microvasc Res ; 123: 42-49, 2019 05.
Article in English | MEDLINE | ID: mdl-30367840

ABSTRACT

Tetrahydroxystilbene glucoside (TSG) is the main water-soluble component in Polygonum multiflorum Thunb, and it has many cardioprotective effects. Although TSG is able to relax blood vessels, its relaxation of rat superior mesenteric arteries and the underlying mechanism of this process are not clearly understood. The aim of the present study was to use in vivo and in vitro models to investigate the arterial relaxation effect of TSG on rat superior mesenteric arteries and the mechanisms involved. We found that TSG concentration-dependently relaxed the superior mesenteric artery with or without endothelium. The vasorelaxation induced by TSG is not related to the vasodilator derived factor NO but is rather by the inhibition of COX-2 activity and decreased TXA2. We also found that the vasorelaxation induced by TSG was attenuated by 4­AP. Moreover, TSG also inhibited the contraction induced by an increase in external calcium concentration in Ca2+-free medium plus KCl (60 mM). These results suggest that TSG induces relaxation in mesenteric arterial rings through an endothelium-dependent pathway that involves the inhibition of COX-2 activity and decreased in TXA2 and through an endothelium-independent pathway via opening of a voltage-dependent K+ channel, blockade of Ca2+ influx and release of intracellular Ca2+.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Glucosides/pharmacology , Muscle, Smooth, Vascular/drug effects , Stilbenes/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Calcium Signaling/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Male , Mesenteric Artery, Superior/drug effects , Mesenteric Artery, Superior/metabolism , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Potassium Channels, Voltage-Gated/agonists , Potassium Channels, Voltage-Gated/metabolism , Rats, Sprague-Dawley , Thromboxane A2/metabolism
12.
Cancer Cell Int ; 18: 195, 2018.
Article in English | MEDLINE | ID: mdl-30524203

ABSTRACT

BACKGROUND: MicroRNAs play crucial roles in tumorigenesis and tumor progression. miR-770 has been reported to be downregulated in several cancers and affects cancer cell proliferation, apoptosis, metastasis and drug resistance. However, the role and underlying molecular mechanism of miR-770 in human glioma remain unknown and need to be further elucidated. METHODS: The expression of miR-770 in glioma tissues and cell lines was measured by quantitative real-time PCR (qRT-PCR) to explore the association of miR-770 expression with clinicopathological characteristics. The expression of CDK8 was detected by qRT-PCR and Western blotting in glioma tissues. A target prediction program and a dual-luciferase reporter assay were used to confirm that CDK8 is a target gene of miR-770. MTT and cell counting assays were used to assess the effect of miR-770 on glioma cell proliferation. The cell cycle distribution and apoptosis were examined by flow cytometry. CDK8 siRNA and overexpression were used to further confirm the function of the target gene. RESULTS: We demonstrated that miR-770 expression was downregulated in human glioma tissues and cell lines. The overexpression of miR-770 inhibited glioma cell proliferation and cell cycle G1-S transition and induced apoptosis. The inhibition of miR-770 facilitated cell proliferation and G1-S transition and suppressed apoptosis. miR-770 expression was inversely correlated with CDK8 expression in glioma tissues. CDK8 was confirmed to be a direct target of miR-770 by using a luciferase reporter assay. The overexpression of miR-770 decreased CDK8 expression at both the mRNA and protein levels, and the suppression of miR-770 increased CDK8 expression. Importantly, CDK8 silencing recapitulated the cellular and molecular effects observed upon miR-770 overexpression, and CDK8 overexpression eliminated the effects of miR-770 overexpression on glioma cells. Moreover, both exogenous expression of miR-770 and silencing of CDK8 resulted in suppression of the Wnt/ß-catenin signaling pathway. CONCLUSIONS: Our study demonstrates that miR-770 inhibits glioma cell proliferation and G1-S transition and induces apoptosis through suppression of the Wnt/ß-catenin signaling pathway by targeting CDK8. These findings suggest that miR-770 plays a significant role in glioma progression and serves as a potential therapeutic target for glioma.

13.
Mol Med Rep ; 18(6): 5229-5236, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272323

ABSTRACT

Cerebral vasospasm (CVS) is a severe complication of subarachnoid hemorrhage (SAH), and endothelin­1 (ET­1) may be involved in its pathogenesis. The present study aimed to investigate the expression of ET­1 in cerebrospinal fluid (CSF) in patients with SAH and to analyze rat arterial contractility and the expression levels of ET­1 receptors in vitro. CSF samples were collected from 28 patients and the expression levels of ET­1 were measured. Rat cerebral basilar arteries were isolated and incubated with hemorrhagic or clear CSF. Contractility, as well as ETA and ETB mRNA expression were measured. ET­1 levels in CSF increased and reached a peak within the initial 5 days after SAH onset and then gradually subsided. After 12 or 24 h, the contraction of arteries incubated in hemorrhagic CSF was substantially stronger than those in clear CSF. The mRNA expression levels of endothelin receptor type A and B in arteries incubated in hemorrhagic CSF were significantly higher than those in clear CSF. ET­1 and its receptors may be involved in the pathogenic mechanism of CVS following SAH. ET­1 expression in CSF may be used as a marker in CVS and its receptors may provide novel therapeutic targets in CVS.


Subject(s)
Endothelin-1/metabolism , Receptor, Endothelin A/metabolism , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/etiology , Vasospasm, Intracranial/physiopathology , Adult , Aged , Animals , Computed Tomography Angiography , Disease Models, Animal , Endothelin-1/genetics , Endothelin-1/pharmacology , Female , Gene Expression , Humans , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Endothelin A/genetics , Subarachnoid Hemorrhage/cerebrospinal fluid , Subarachnoid Hemorrhage/diagnosis , Time Factors , Tomography, X-Ray Computed
14.
Adv Pharmacol Sci ; 2017: 6507048, 2017.
Article in English | MEDLINE | ID: mdl-28775740

ABSTRACT

Thromboxane A2 (TXA2) acts on TXA2 receptors (TP) to regulate renal artery blood flow and subsequently contributes to the pathogenesis of renal ischemia. The present study was designed to examine if nuclear factor-kappaB (NF-κB) signaling pathway is involved in the downregulation of TP receptors in rat renal artery. Rat renal artery segments were organ cultured for 6 or 24 h. Downregulation of TP receptors was monitored using myograph (contractile function), real-time PCR (receptor mRNA), and immunohistochemistry (receptor protein). Specific inhibitors (MG-132 and BMS345541) for NF-κB signaling pathway were used to dissect the underlying molecular mechanisms involved. Compared to fresh (noncultured) segments, organ culture of the renal artery segments for 24 h induced a significant rightward shift of U46619 (TP receptor agonist) contractile response curves (pEC50: 6.89 ± 0.06 versus 6.48 ± 0.04, P < 0.001). This decreased contractile response to U46619 was paralleled with decreased TP receptor mRNA and protein expressions in the renal artery smooth muscle cells. Specific inhibitors (MG-132 and BMS345541) for NF-κB signaling pathway significantly abolished the decreased TP protein expression and receptor-mediated contractions. In conclusion, downregulation of TP receptors in the renal artery smooth muscle cells occurs mainly via the NF-κB signaling pathway.

15.
Vascul Pharmacol ; 96-98: 33-39, 2017 09.
Article in English | MEDLINE | ID: mdl-28797761

ABSTRACT

Minimally modified low density lipoprotein (mmLDL) is a risk factor for cardiovascular disease. This study investigated the effect of mmLDL on mouse mesenteric artery endothelin type B (ETB) receptors and its molecular mechanism. Mice were injected with normal saline (NS group), mmLDL in the tail vein (mmLDL group), or with both mmLDL and an intraperitoneal injection of the ERK1/2 pathway-specific inhibitor U0126 (mmLDL+U0126 group). The dose-response curve of mesenteric artery contraction induced by sarafotoxin 6c (S6c), the ETB receptor agonist, was measured using a sensitive myograph system. ELISAs, RT-PCR and Western blot were used to determine the serum concentrations of mouse oxidized low density lipoprotein (oxLDL), intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) as well as the expression of ETB receptors, ICAM-1, VCAM-1 and phosphorylated-extracellular signal-regulated kinase 1/2 (p-ERK1/2). The S6c-induced contraction dose-response curve was significantly enhanced by mmLDL treatment and showed a significantly higher Emax value than in the NS group (P<0.001), and the ETB receptor mRNA and protein expression in the vascular wall was significantly higher than in the NS group. The serum concentration and expression of ICAM-1 and VCAM-1 were also increased by mmLDL treatment, but intraperitoneal injection of U0126 inhibited these changes as well as the increase in p-ERK1/2 protein in the vessel wall caused by mmLDL. ICAM-1 and VCAM-1 serum concentrations were positively correlated with the S6c-induced maximum contraction of blood vessels. Increased in vivo levels of mmLDL increased the serum concentrations and expression of ICAM-1 and VCAM-1 by activating the ERK1/2 pathway, resulting in the expression of ETB receptors and the enhancement of contractile function in vascular smooth muscle. Understanding the effect of mmLDL on ETB receptors and its mechanism can provide ideas for cardiovascular disease prevention and treatment.


Subject(s)
Lipoproteins, LDL/administration & dosage , Mesenteric Arteries/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, Endothelin B/drug effects , Signal Transduction/drug effects , Vasoconstriction/drug effects , Animals , Butadienes/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Female , In Vitro Techniques , Injections, Intravenous , Intercellular Adhesion Molecule-1/metabolism , Male , Mesenteric Arteries/enzymology , Mice, Inbred ICR , Nitriles/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptor, Endothelin B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vasoconstrictor Agents/pharmacology , Viper Venoms/pharmacology
16.
Front Pharmacol ; 8: 372, 2017.
Article in English | MEDLINE | ID: mdl-28659805

ABSTRACT

Numerous studies have noted that populations treated with statins have increased risk for new-onset diabetes mellitus; however, the underlying molecular mechanisms are not fully understood. Interestingly, familial hypercholesterolemia (FH) patients with mutations in the low-density lipoprotein receptor (LDLR) gene are protected against diabetes mellitus (DM), despite these patients being subjected to long-term statin therapy. Since the common pathway between FH and statin therapy is LDLR-mediated cellular cholesterol uptake, the arising question is whether the LDLR plays an important role in the diabetogenic effect of statins. Indeed, given that statins can regulate the LDLR expression in liver and peripheral tissue, there is a possible mechanism that the increased LDLR causes cellular cholesterol accumulation and dysfunction in pancreatic islets, explaining why statins fail to increase the risk of DM in FH patients. In this paper, with regarded to recent literatures, we highlight the role of LDLR in the pathophysiology of cholesterol-induced pancreatic islets dysfunction, which may provide the key link between statins treatment and the increased risk of new-onset diabetes mellitus.

17.
Basic Clin Pharmacol Toxicol ; 120(4): 327-334, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27731935

ABSTRACT

Abnormal contraction of vessels termed 'vasospasm' is associated with various cardiovascular diseases. Smoking is a well-known risk factor that increases vasospasm. However, the molecular mechanisms by which smoking leads to vasospasm and cardiovascular disease are not fully understood. This study was designed to examine whether DMSO-extracted cigarette smoke particles (DSP) could induce up-regulation of vascular endothelin type A (ETA ) receptors, and whether ETA receptor is up-regulated through activation of extracellular regulated protein kinases 1 and 2 (ERK1/2) signal pathways. Mesenteric arterial segments from rats were cultured in the presence of DSP, water-extracted cigarette smoke particles (WSP) or equivalent concentration of nicotine for up to 24 hr. The results showed that DSP, but not WSP or nicotine, induced ETA receptor up-regulation with increased ETA receptor-mediated contraction (myograph, p < 0.001). Simultaneously, the expression of ETA receptor mRNA (real-time PCR, p < 0.001) and protein (immunohistochemistry) were enhanced in the smooth muscle cells, suggesting that the lipid-soluble substances contained in cigarette smoke were responsible for the effects of DSP. Actinomycin D (a general transcriptional inhibitor) decreased ETA receptor mRNA expression and attenuated receptor-mediated contraction (p < 0.001), while DSP accelerated ETA receptor mRNA degradation (p < 0.01) and promoted the translation of ETA receptor mRNA into protein. Furthermore, the up-regulation of ETA receptors was significantly attenuated by inhibition of ERK1/2 signal pathways (p < 0.001). In conclusion, DSP most likely activate ERK1/2 signal pathway-mediated transcriptional and post-transcriptional (translational) mechanisms that lead to vascular ETA receptor up-regulation, which might contribute to vasospasm and the development of smoking-associated cardiovascular diseases.


Subject(s)
Lipids/chemistry , MAP Kinase Signaling System/drug effects , Mesenteric Artery, Superior/drug effects , Particulate Matter/toxicity , Receptor, Endothelin A/genetics , Tobacco Smoke Pollution/adverse effects , Animals , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Immunohistochemistry , In Vitro Techniques , Male , Mesenteric Artery, Superior/metabolism , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction , Solubility , Up-Regulation
18.
Biomed Pharmacother ; 84: 1979-1985, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27847206

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for cardiovascular diseases (CVDs). Endothelin type B (ETB) receptors were involved in the pathogenesis of CVDs. However, Sirtuin 1(Sirt1) has potential protect roles for CVDs. The present study was designed to examine the hypothesis that homocysteine up-regulates ETB receptors through down-regulation of Sirt 1. In vitro experiments were performed in rat superior mesenteric artery (SMA). The rat SMA was cultured in serum free medium for 24h in the presence and absence of homocysteine (Hcy) with or without resveroral (Res) (a Sirt 1agonist). In vivo, the rats received subcutaneous injections of Hcy in the presence of or absence of Res for 3 weeks. The contractile responses to sarafotoxin 6c (S6c) (an ETB receptor agonist) were studied using a sensitive myograph. Levels of protein expression were determined using western blotting. The blood pressure of rat was measured via a noninvasive tail-cuff plethysmography method. We observed that Hcy increased the level of ETB receptor protein expression and the ETB receptor-mediated contractile responses induced by S6c, and decreased level of Sirt1 protein expression in SMA without endothelium in vitro. However, these effects were reversed by Res. Moreover, Res also blocked the up-regulation of acetylized p65 induced by Hcy. The in vivo study showed that HHcy down-regulated Sirt 1, and up-regulated acetylized p65 and ETB receptor protein expression, and elevated the blood pressure of rats. However, Res could block these effects. In conclusion, this suggested that Hcy regulated ETB receptor expression through sirt1/nuclear factor-κB signaling pathway.


Subject(s)
Homocysteine/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Receptor, Endothelin B/physiology , Sirtuin 1/metabolism , Animals , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Sirtuin 1/antagonists & inhibitors
19.
J Vasc Res ; 53(1-2): 58-71, 2016.
Article in English | MEDLINE | ID: mdl-27577886

ABSTRACT

Minimally modified low-density lipoprotein (mmLDL) is a well-known risk factor for cardiovascular diseases. The present study was designed to investigate the role of mmLDL in the endothelium-dependent relaxation of mouse mesenteric arteries. A sensitive myograph system was employed to examine the endothelial function of mesenteric arteries. mRNA and protein expression levels were determined using real-time PCR and Western blotting, respectively. The ultramicrostructure of mesenteric vascular beds was investigated using a transmission electron microscope. The results showed that mmLDL significantly impaired the acetylcholine-induced (3 × 10-10 to 1 × 10-4M) endothelium-dependent relaxation of mouse mesenteric arteries with markedly reduced pIC50 (p < 0.05) and Rmax values (p < 0.001). In addition, mmLDL increased the levels of superoxide production and nitrotyrosine concentration and impaired the endothelial microstructure with decreased KCa3.1 and KCa2.3 expression. In conclusion, mmLDL increases superoxide and nitrotyrosine levels, damages endothelial microstructure with decreased KCa3.1 and KCa2.3 expression, and ultimately attenuates relaxation mediated by nitric oxide- and endothelium-derived hyperpolarizing factor.


Subject(s)
Endothelium, Vascular/drug effects , Lipoproteins, LDL/pharmacology , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Animals , Biological Factors/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Gene Expression Regulation , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/ultrastructure , Mice, Inbred ICR , Nitric Oxide/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Superoxides/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
20.
Vascul Pharmacol ; 87: 100-109, 2016 12.
Article in English | MEDLINE | ID: mdl-27592458

ABSTRACT

Vascular smooth muscle endothelin type B (ETB) receptor is involved in the pathogenesis of cardiovascular diseases (CVDs). Hyperhomocysteinemia is an independent risk factor for CVDs. The present study was designed to examine the hypothesis that homocysteine (Hcy) up-regulates vascular smooth muscle ETB receptors. In vitro experiments were performed in rat superior mesenteric artery (SMA) and vascular smooth muscle cells (VSMCs). The rat SMA or VSMCs were cultured in serum-free medium for 24h in the presence and absence of Hcy with or without specific inhibitors for the ERK1/2 signaling pathway and NF-κB. In vivo, the rats received subcutaneous injections of Hcy in the presence or absence of specific inhibitors for the ERK1/2 signaling pathway (U0126) for 3weeks. Levels of protein expression were determined using Western blot analysis. The contractile responses to sarafotoxin 6c (an ETB receptor agonist) were studied using a sensitive myograph. The blood pressure of the rats was measured via a noninvasive tail-cuff plethysmography method. The results from in vitro experiments showed that Hcy concentration-dependently increased the ETB receptor-mediated contractile responses, and up-regulated ETB receptor expression, in rat SMA. Blockage of the ERK1/2 signaling pathway and NF-κB using the MEK1/2 inhibitor (PD98059 and U0126) or IκB kinase inhibitor (wedelolactone) significantly abolished Hcy-induced up-regulation of ETB receptor. Finally, we used VSMCs as a cellular model to further validate our finding. In vivo study found that hyperhomocysteinemia up-regulated ETB receptor expression, and elevated the blood pressure of rats via the ERK1/2 signaling pathway. In conclusion, Hcy up-regulated vascular smooth muscle ETB receptor via activation of the ERK1/2 signaling pathway and NF-κB.


Subject(s)
Homocysteine/metabolism , Hyperhomocysteinemia/complications , Muscle, Smooth, Vascular/metabolism , Receptor, Endothelin B/metabolism , Animals , Blood Pressure , Blotting, Western , MAP Kinase Signaling System , Male , Mesenteric Artery, Superior/metabolism , Muscle, Smooth, Vascular/cytology , NF-kappa B/metabolism , Plethysmography/methods , Rats , Rats, Sprague-Dawley , Receptor, Endothelin B/genetics , Up-Regulation , Viper Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...