Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cardiovasc Diabetol ; 23(1): 274, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39049097

RESUMO

Diabetes mellitus (DM) is a metabolic disease that heightens the risks of many vascular complications, including peripheral arterial disease (PAD). Various types of cells, including but not limited to endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and macrophages (MΦs), play crucial roles in the pathogenesis of DM-PAD. Long non-coding RNAs (lncRNAs) are epigenetic regulators that play important roles in cellular function, and their dysregulation in DM can contribute to PAD. This review focuses on the developing field of lncRNAs and their emerging roles in linking DM and PAD. We review the studies investigating the role of lncRNAs in crucial cellular processes contributing to DM-PAD, including those in ECs, VSMCs, and MΦ. By examining the intricate molecular landscape governed by lncRNAs in these relevant cell types, we hope to shed light on the roles of lncRNAs in EC dysfunction, inflammatory responses, and vascular remodeling contributing to DM-PAD. Additionally, we provide an overview of the research approach and methodologies, from identifying disease-relevant lncRNAs to characterizing their molecular and cellular functions in the context of DM-PAD. We also discuss the potential of leveraging lncRNAs in the diagnosis and therapeutics for DM-PAD. Collectively, this review provides a summary of lncRNA-regulated cell functions contributing to DM-PAD and highlights the translational potential of leveraging lncRNA biology to tackle this increasingly prevalent and complex disease.


Assuntos
Células Endoteliais , Macrófagos , Miócitos de Músculo Liso , Doença Arterial Periférica , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Doença Arterial Periférica/genética , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/fisiopatologia , Animais , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Regulação da Expressão Gênica , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/fisiopatologia , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/diagnóstico , Transdução de Sinais , Remodelação Vascular/genética , Epigênese Genética
2.
Vasc Med ; 28(1): 6-17, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36759934

RESUMO

BACKGROUND: Circular RNAs (circRNAs) have been implicated in the pathogenesis of atherosclerosis (AS) and the migration and proliferation of vascular smooth muscle cells (VSMCs) under oxidized low-density lipoprotein (ox-LDL). Here, we defined the exact action of human circ_0007478 in VSMC migration and proliferation induced by ox-LDL. METHODS: Human VSMCs (HVSMCs) were exposed to ox-LDL. Circ_0007478, microRNA (miR)-638, and rho-associated protein kinase 2 (ROCK2) levels were gauged by quantitative real-time PCR (qRT-PCR) and western blot. Cell viability and proliferation were assessed by MTT and EdU assays, respectively. Transwell assays were used to detect cell migration and invasion. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were used to evaluate the direct relationship between miR-638 and circ_0007478 or ROCK2. RESULTS: Our data indicated that circ_0007478 expression was augmented in AS serum samples and ox-LDL-treated HVSMCs. Depletion of circ_0007478 attenuated HVSMC proliferation, migration, and invasion induced by ox-LDL. Mechanistically, circ_0007478 targeted miR-638 by directly pairing to miR-638. Reduction of miR-638 reversed the effects of circ_0007478 depletion on ox-LDL-evoked proliferation, migration, and invasion in HVSMCs. ROCK2 was a direct miR-638 target and miR-638-mediated inhibition of ROCK2 relieved ox-LDL-evoked HVSMC proliferation, migration, and invasion. Furthermore, circ_0007478 was identified as a competing endogenous RNA (ceRNA) for miR-638 to modulate ROCK2 expression. CONCLUSION: Our present study establishes an undescribed ceRNA regulatory network, in which circ_0007478 targets miR-638 to upregulate ROCK2, thereby contributing to ox-LDL-induced proliferation and migration in HVSMCs.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Músculo Liso Vascular , Aterosclerose/genética , Movimento Celular , Lipoproteínas LDL/farmacologia , Proliferação de Células , MicroRNAs/genética , Apoptose , Células Cultivadas , Miócitos de Músculo Liso , Quinases Associadas a rho/genética
3.
Biol Chem ; 403(3): 317-329, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34882999

RESUMO

Abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by insulin resistance facilitates intimal hyperplasia of type 2 diabetes mellitus (T2DM) and N6-methyladenosine (m6A) methylation modification mediates the VSMC proliferation. This study aimed to reveal the m6A methylation modification regulatory mechanism. In this study, m6A demethylase FTO was elevated in insulin-treated VSMCs and T2DM mice with intimal injury. Functionally, FTO knockdown elevated m6A methylation level and further restrained VSMC proliferation and migration induced by insulin. Mechanistically, FTO knockdown elevated Smooth muscle 22 alpha (SM22α) expression and m6A-binding protein IGF2BP2 enhanced SM22α mRNA stability by recognizing and binding to m6A methylation modified mRNA. In vivo studies confirmed that the elevated m6A modification level of SM22α mRNA mitigated intimal hyperplasia in T2DM mice. Conclusively, m6A methylation-mediated elevation of SM22α restrained VSMC proliferation and migration and ameliorated intimal hyperplasia in T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Animais , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Insulinas/metabolismo , Metilação , Camundongos , Músculo Liso Vascular/patologia , RNA Mensageiro/metabolismo
4.
Zhongguo Zhong Yao Za Zhi ; 46(17): 4497-4503, 2021 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-34581055

RESUMO

This study aimed to observe the inhibitory effect of icariin against oxidative stress-induced calcification in aortic vascular smooth muscle cells(VSMCs) and elucidate the molecular mechanism of icariin in inhibiting endoplasmic reticulum stress(ERS)-mediated atherosclerotic calcification, so as to provide new ideas for exploring the anti-atherosclerotic mechanism of Epimedii Folium. The VSMCs in rat thoracic aorta were subjected to adherent culture and then treated with the complete calcification DMEM containing high glucose and hydrogen peroxide(H_2O_2) for three weeks. The resulting calcified VSMCs were divided into different treatment groups. Icariin was added one week after calcification induction for protecting the VSMCs, whose viability was then detected using cell counting kit-8(CCK-8). Alizarin red-S staining was conducted to observe the calcification degree. The activity of alkaline phosphatase(ALP) in VSMCs was measured using the disodium phenyl phosphate substrate and the calcium content was measured by arsenazo Ⅲ method. The mRNA expression levels of ossification-related factors including osteocalcin(OC), osteopontin(OPN), Runt-related transcription factor 2(Runx2), and type Ⅰ collagen(Col Ⅰa) were detected by real-time PCR. Western blot was carried out to determine the protein expression levels of α-smooth muscle actin(α-SMA), Runx2, activating transcription factor 4(ATF4), and eukaryotic translation initiation factor(eIF)-2α. The results showed that H_2O_2 significantly induced the calcification of VSMCs, increased the ALP activity and calcium content in VSMCs, promoted OC, OPN, Runx2, and Col Ⅰa mRNA expression and Runx2 protein expression, and reduced α-SMA protein expression. The ATF4 protein expression and eIF2α phosphorylation were also elevated significantly. Icariin reversed the calcification of VSMCs induced by H_2O_2, inhibited ALP activity and calcium content in VSMCs, down-regulated the mRNA expression levels of OC, OPN, Runx2 and Col Ⅰa and Runx2 protein expression, and relatively up-regulated the expression of α-SMA. The expression of ATF4 and phosphorylation of eIF2α also declined significantly. All these have demonstrated that icariin inhibited VSMCs calcification by down-regulating the ossification-related factors and lowering ALP activity and calcium content in VSMCs. Besides, the down-regulation of Runx2 expression and the inhibition of ATF4 and eIF2α-mediated cellular calcification pathway in ERS might also be involved in such calcification-suppressing process.


Assuntos
Músculo Liso Vascular , Miócitos de Músculo Liso , Animais , Células Cultivadas , Flavonoides/metabolismo , Flavonoides/farmacologia , Músculo Liso Vascular/metabolismo , Estresse Oxidativo , Ratos
5.
Biochem Cell Biol ; 98(2): 249-257, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32207314

RESUMO

In this study we investigated the regulatory role of cell-migration-inducing and hyaluronan-binding protein (CEMIP) in the proliferation and migration of vascular smooth muscle cells (VSMCs). The mRNA and protein levels of CEMIP were upregulated in the plasma samples from patients with atherosclerosis, and in VSMCs stimulated with platelet-derived growth factor-BB (PDGF-BB), compared with plasma from healthy subjects and untreated VSMCs. Silencing CEMIP suppressed PDGF-BB-induced cell migration and proliferation in VSMCs, as determined using a Cell Counting Kit-8 assays, 5-ethynyl-2'-deocyuridine (EDU) assays, flow cytometry, wound healing assays, and Transwell assays. Overexpression of CEMIP promoted the proliferation and migration of VSMCs via activation of the Wnt-ß-catenin signaling pathway and the upregulation of its target genes, including matrix metalloproteinase-2, matrix metalloproteinase-7, cyclin D1, and c-myc, whereas CEMIP deficiency showed the opposite effects. The knockdown of CEMIP in ApoE-/- mice by intravenous injection of lentiviral vector expressing si-CEMIP protected against high-fat-diet-induced atherosclerosis, as shown by the reduced aortic lesion areas, aortic sinus lesion areas, and the concentration of blood lipids compared with mice normally expressing CEMIP. These results demonstrated that CEMIP regulates the proliferation and migration of VSMCs in atherosclerosis by activating the WNT-ß-catenin signaling pathway, which suggests the therapeutic potential of CEMIP for the management of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Hialuronoglucosaminidase/metabolismo , Miócitos de Músculo Liso/citologia , Via de Sinalização Wnt , Animais , Becaplermina/metabolismo , Movimento Celular , Proliferação de Células , Feminino , Citometria de Fluxo , Inativação Gênica , Humanos , Lentivirus/metabolismo , Lipídeos/sangue , Masculino , Camundongos , Camundongos Knockout para ApoE , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , RNA Interferente Pequeno/metabolismo , Regulação para Cima , Cicatrização
6.
Circ J ; 84(12): 2259-2269, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33162460

RESUMO

BACKGROUND: The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the development of atherosclerosis. This study aimed to investigate the role of circular RNA-0010283 (circ_0010283) in oxidized low-density lipoprotein (ox-LDL)-treated VSMCs and the associated action mechanism.Methods and Results:The expression of circ_0010283 was investigated using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was monitored by using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis was detected by using flow cytometry assay. A transwell assay was performed to observe migration and invasion, and a scratch assay was implemented to test migration. The expression of proliferation, apoptosis and migration/invasion-related proteins was measured by using a western blot. The targeted relationship was predicted by using a bioinformatics tool (Starbase) and verified by using a dual-luciferase reporter assay, a RNA immunoprecipitation (RIP) assay and a RNA pull-down assay. circ_0010283 was highly expressed in serum samples from atherosclerosis patients and ox-LDL-treated human VSMCs (HVSMCs). circ_0010283 knockdown suppressed ox-LDL-induced proliferation, migration and invasion in HVSMCs. MicroRNA-133a-3p (miR-133a-3p) was confirmed as a target of circ_0010283, and miR-133a-3p deficiency reversed the effects of circ_0010283 knockdown. Moreover, pregnancy-associated plasma protein A (PAPPA) was targeted by miR-133a-3p, and PAPPA overexpression reversed the effects of miR-133a-3p restoration. Interestingly, circ_0010283 could regulate PAPPA expression by mediating miR-133a-3p. CONCLUSIONS: circ_0010283 participated in ox-LDL-induced dysfunctions of HVSMCs by modulating the miR-133a-3p/PAPPA pathway, suggesting that circ_0010283 might be associated with atherosclerosis pathogenesis.


Assuntos
Aterosclerose , Lipoproteínas LDL/metabolismo , MicroRNAs , Miócitos de Músculo Liso/citologia , Proteína Plasmática A Associada à Gravidez/genética , RNA Circular , Apoptose , Aterosclerose/genética , Proteínas Sanguíneas , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , MicroRNAs/genética , Músculo Liso Vascular/citologia , RNA Circular/genética
7.
Acta Biochim Biophys Sin (Shanghai) ; 52(9): 935-943, 2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32785574

RESUMO

Homeobox c6 (Hoxc6) affects the proliferation, migration, and infiltration of malignant tumor cells; however, the effect of Hoxc6 on atherosclerosis (AS) as well as the proliferation and migration of vascular smooth muscle cells (VSMCs), which play a role in promoting AS, has not yet been well clarified. In the present study, we tested the hypothesis that Hoxc6 affects the proliferation and migration of rat VSMCs, and hence is involved in AS. The results showed that the expression of Hoxc6 mRNA and protein was higher in normal rat aortic wall than in the myocardium. Subsequently, a rat model of AS was established by high-fat feeding for 2 months. The expression of Hoxc6 mRNA and protein was increased significantly in AS lesions, while the expression of p53 protein was decreased and that of proliferating cell nuclear antigen (PCNA) was increased. Moreover, not only the proliferation and mobility of cells in normal culture were decreased, but also the proliferation was stimulated by oxidized low-density lipoprotein, which was decreased after downregulation of Hoxc6 expression in VSMCs in rat. Consecutively, the expression of PCNA protein was decreased, while that of p53 was increased. These results indicated that Hoxc6 is probably involved in AS via p53 and PCNA by affecting the proliferation and migration of VSMCs.


Assuntos
Aorta/metabolismo , Aterosclerose/metabolismo , Movimento Celular , Proliferação de Células , Regulação da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Aorta/patologia , Aterosclerose/patologia , Masculino , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Ratos , Ratos Wistar
8.
Cell Biol Int ; 43(10): 1102-1112, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31066128

RESUMO

MicroRNAs (miRNAs) are a class of nonprotein-encoding RNAs of ~22 nucleotides in length that bind to or complement each other with a target gene messenger RNA (mRNA) to promote mRNA degradation or inhibit translation of the target mRNA. The protein required [such as Toll-like receptor (TLR) proteins] is controlled at an optimal level. By affecting protein translation, miRNAs have become powerful regulators of biological processes, including development, differentiation, cell proliferation, and apoptosis. MiRNAs are involved in the regulation of proliferation, migration, and apoptosis of vascular smooth muscle cells (VSMCs), thereby affecting the formation of atherosclerosis (AS). In recent years, the role and mechanism of miRNAs involved in AS development in VSMCs have been studied extensively. In the current study, the results and progress in miRNA research are reviewed.


Assuntos
Aterosclerose/metabolismo , MicroRNAs/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Apoptose , Proliferação de Células , Humanos , Camundongos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Ratos
9.
Mol Cell Biochem ; 439(1-2): 131-140, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28871467

RESUMO

Epigallocatechin gallate (EGCG), a bioactive ingredient of green tea, plays a protective role in the cardiovascular system. Homocysteine (Hcy) is a major risk factor for chronic kidney disease and cardiovascular disease. The present study aimed to investigate the role of EGCG in Hcy-induced proliferation of vascular smooth muscle cells (VSMCs) and its underlying mechanism. We also explored the roles of rennin-angiotensin system (RAS), extracellular signal-regulated kinases (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK) in this process. Human aortic smooth muscle cells (HASMCs) were treated with different drugs for different periods. The proliferation rate of HASMCs was detected using the CCK-8 and BrdU labeling assays. The Western blot assay was used to determine the expression levels of angiotensin II type 1 receptor (AT-1R), ERK1/2, and p38 MAPK. Compared with the control group, the HASMCs treated with Hcy at different doses (100, 200, 500, and 1000 µM) showed significantly increased proliferation. Hcy increased the expression of AT-1R, whereas EGCG decreased the protein expression of AT-1R. Furthermore, we found that Hcy-induced expression of p-ERK1/2 and p-p38MAPK was dependent on AT-1R. Compared with Hcy (500 µM)-treated cells, EGCG (20 µM)-treated cells showed decreased proliferation as well as expression of AT-1R, p-ERK1/2, and p-p38MAPK. In addition, HASMC proliferation was suppressed by the addition of an AT-1R blocker (olmesartan), an ERK1/2 inhibitor (PD98059), and a p38MAPK inhibitor (SB202190). EGCG can inhibit AT-1R and affect ERK1/2 and p38MAPK signaling pathways, resulting in the decrease of VSMC proliferation induced by Hcy.


Assuntos
Catequina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Homocisteína/efeitos adversos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Catequina/farmacologia , Linhagem Celular , Homocisteína/farmacologia , Humanos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia
10.
Biochem Biophys Res Commun ; 485(1): 215-220, 2017 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-28193524

RESUMO

Apoptosis plays a critical role in normal vascular development and atherosclerosis. However, high glucose has been reported to generate a certain level of ROS that can inhibit vascular smooth muscle cell (VSMC) apoptosis, with the underlying mechanism remaining unclear. In this study, a synthetic peptide AREGEM (Ala-Arg-Glu-Gly-Glu-Met) exhibited antioxidative effects and was used to investigate its function in VSMCs during hyperglycaemia. MTT assay results demonstrated that AREGEM significantly attenuated high glucose-induced VSMCs proliferation. Flow cytometry displayed that high glucose levels inhibited cell apoptosis, whereas this effect was attenuated by pre-incubation with AREGEM. In addition, the 2',7'-dichlorofluorescein diacetate (DCFH-DA) fluorescent probe assay further demonstrated that AREGEM reduced intracellular ROS accumulation in VSMCs. Furthermore, this peptide was able to prevent the decrease of caspase-3 activity and the increase of the ratio of Bcl-2/Bax protein in VSMCs exposed to high glucose. These findings demonstrated that AREGEM is able to abolish the effects of high glucose in VSMCs; therefore, this peptide can be a potential candidate to develop a novel strategy for curing diabetic related diseases.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glucose/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Peptídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sequência de Aminoácidos , Antioxidantes/química , Caspase 3/metabolismo , Linhagem Celular , Humanos , Hiperglicemia/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Peptídeos/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
11.
Biochem Biophys Res Commun ; 487(3): 587-593, 2017 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-28433630

RESUMO

Abdominal aortic aneurysm (AAA) is relatively common in elderly patients with atherosclerosis. MURC (muscle-restricted coiled-coil protein)/Cavin-4 modulating the caveolae function of muscle cells is expressed in cardiomyocytes, skeletal muscle cells and smooth muscle cells. Here, we show a novel functional role of MURC/Cavin-4 in vascular smooth muscle cells (VSMCs) and AAA development. Both wild-type (WT) and MURC/Cavin-4 knockout (MURC-/-) mice subjected to periaortic application of CaCl2 developed AAAs. Six weeks after CaCl2 treatment, internal and external aortic diameters were significantly increased in MURC-/- AAAs compared with WT AAAs, which were accompanied by advanced fibrosis in the tunica media of MURC-/- AAAs. The activity of JNK and matrix metalloproteinase (MMP) -2 and -9 were increased in MURC-/- AAAs compared with WT AAAs at 5 days after CaCl2 treatment. At 6 weeks after CaCl2 treatment, MURC-/- AAAs exhibited attenuated JNK activity compared with WT AAAs. There was no difference in the activity of MMP-2 or -9 between saline and CaCl2 treatments. In MURC/Cavin-4-knockdown VSMCs, TNFα-induced activity of JNK and MMP-9 was enhanced compared with control VSMCs. Furthermore, WT, MURC-/-, apolipoprotein E-/- (ApoE-/-), and MURC/Cavin-4 and ApoE double-knockout (MURC-/-ApoE-/-) mice were subjected to angiotensin II (Ang II) infusion. In both ApoE-/- and MURC-/-ApoE-/- mice infused for 4 weeks with Ang II, AAAs were promoted. The internal aortic diameter was significantly increased in Ang II-infused MURC-/-ApoE-/- mice compared with Ang II-infused ApoE-/- mice. In MURC/Cavin-4-knockdown VSMCs, Ang II-induced activity of JNK and MMP-9 was enhanced compared with control VSMCs. Our results suggest that MURC/Cavin-4 in VSMCs modulates AAA progression at the early stage via the activation of JNK and MMP-9. MURC/Cavin-4 is a potential therapeutic target against AAA progression.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Musculares/deficiência , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Animais , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia
12.
Cell Tissue Res ; 370(3): 365-377, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28803422

RESUMO

Vessel disease is a kind of severe complication in diabetic patients. However, few pharmacologic agents can directly recover diabetic vascular function. Salidroside (SAL), a major ingredient from Rhodiola rosea, has been found to have an obvious hypoglycemic effect and a beneficial protection on vascular function in diabetes. However, whether SAL is a suitable treatment for diabetes has not so far been evaluated and the underlying mechanisms remain unknown. The present work aims to (1) investigate the potential effects of SAL on cerebrovascular relaxation in streptozotocin-induced diabetic rats or when exposed to acute hyperglycemia condition and (2) examine whether function of the BKCa channel is involved in SAL treatment for diabetic vascular relaxation. Our results indicate that chronic administration of 100 mg/kg/day SAL not only improves cerebrovascular relaxation but also increases BKCa ß1-subunit expressions at both protein and mRNA levels and enhances BKCa whole-cell and single-channel activities in cerebral VSMCs of diabetic rats. Correspondingly, acute application of 100 µM SAL induces cerebrovascular relaxation by activation of the BKCa channel. Furthermore, SAL activated the BKCa channel mainly through acting on the ß1-subunit in HEK293 cells transfected with hSloα+ß1 constructs. We concluded that SAL improved vasodilation in diabetic rats through restoring the function of the BKCa-ß1 subunit in cerebrovascular smooth muscle cells, which may be the underlying mechanism responsible for the vascular protection of SAL in diabetes.


Assuntos
Glucosídeos/farmacologia , Hipoglicemiantes/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fenóis/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Linhagem Celular , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Células HEK293 , Humanos , Masculino , Miócitos de Músculo Liso/metabolismo , Ratos , Ratos Wistar , Estreptozocina
13.
Mol Cell Biochem ; 433(1-2): 205-211, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28386846

RESUMO

The proliferation-promoting effect of neuropeptide Y (NPY) always functions in low-serum-cultured vascular smooth muscle cells (VSMCs), and the phenotypic switch of VSMCs is regulated by concentrations of serum. Whether the property of the NPY proliferative effect in VSMCs relies on phenotype of VSMCs is unclear. We aimed to explore the role of NPY on proliferation of different VSMC phenotypes in the pathogenesis of atherosclerosis. By stimulating A10 cells with 200 nM NPY in 0.5 or 10% serum, 3H-thymidine and 5-ethynyl-2'-deoxyuridine (EdU) and CCK8 measurements were used to detect VSMC proliferation. RT-PCR and Flow cytometry were performed to detect the factors involved in different properties of the NPY proliferative effect in VSMCs. Instead of facilitating proliferation, NPY had no significant effect on the growth of VSMCs when cultured in 10% serum (VSMCs stayed at synthetic states). The underlying mechanism may be involved in down-regulation of Y1 receptor (P < 0.05 vs. Vehicle) and up-regulation of Geminin (P < 0.05 vs. Vehicle) in 10% serum-cultured VSMCs co-incubated with 200 nM NPY. Besides, modulation of Geminin was effectively blocked by the Y1 receptor antagonist. The stimulation of NPY on proliferation of VSMCs could be a double-edged sword in the development of atherosclerosis and thus provides new knowledge for therapy of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Proliferação de Células/efeitos dos fármacos , Geminina/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neuropeptídeo Y/farmacologia , Animais , Aterosclerose/patologia , Linhagem Celular , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Ratos
14.
Cardiovasc Diabetol ; 15: 63, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27067643

RESUMO

BACKGROUND: Vascular dysfunction is a distinctive phenotype in diabetes mellitus. Current treatments mostly focus on the tight glycemic control and few of these treatments have been designed to directly recover the vascular dysfunction in diabetes. As a classical natural medicine, berberine has been explored as a possible therapy for DM. In addition, it is reported that berberine has an extra-protective effect in diabetic vascular dysfunction. However, little is known whether the berberine treatment could ameliorate the smooth muscle contractility independent of a functional endothelium under hyperglycemia. Furthermore, it remains unknown whether berberine affects the arterial contractility by regulating the intracellular Ca(2+) handling in vascular smooth cells (VSMCs) under hyperglycemia. METHODS: Sprague-Dawley rats were used to establish the diabetic model with a high-fat diet plus injections of streptozotocin (STZ). Berberine (50, 100, and 200 mg/kg/day) were intragastrically administered to control and diabetic rats for 8 weeks since the injection of STZ. The intracellular Ca(2+) handling of isolated cerebral VSMCs was investigated by recording the whole-cell L-type Ca(2+) channel (CaL) currents, assessing the protein expressions of CaL channel, and measuring the intracellular Ca(2+) in response to caffeine. Our results showed that chronic administration of 100 mg/kg/day berberine not only reduced glucose levels, but also inhibited the augmented contractile function of cerebral artery to KCl and 5-hydroxytryptamine (5-HT) in diabetic rats. Furthermore, chronic administration of 100 mg/kg/day berberine significantly inhibited the CaL channel current densities, reduced the α1C-subunit expressions of CaL channel, decreased the resting intracellular Ca(2+) ([Ca(2+)]i) level, and suppressed the Ca(2+) releases from RyRs in cerebral VSMCs isolated from diabetic rats. Correspondingly, acute application of 10 µM berberine could directly inhibit the hyperglycemia-induced CaL currents and suppress the hyperglycemia-induced Ca(2+) releases from RyRs in cerebral VSMCs isolated from normal control rats. CONCLUSIONS: Our study indicated that berberine alleviated the cerebral arterial contractility in the rat model of streptozotocin-induced diabetes via regulating the intracellular Ca(2+) handling of smooth muscle cells.


Assuntos
Berberina/farmacologia , Cálcio/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Masculino , Miócitos de Músculo Liso/metabolismo , Ratos Sprague-Dawley
15.
Biochem Biophys Res Commun ; 460(3): 497-503, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25824048

RESUMO

Endothelin-1 (ET-1) is known as potent vasoconstrictor, by virtue of its mitogenic effects, and may deteriorate the process of hypertension and atherosclerosis by aggravating hyperplasia and migration in VSMCs. Our previous study demonstrated that insulin infusion caused sequential induction of hyperinsulinemia, hyperendothelinemia, insulin resistance, and then hypertension in rats. However, the underlying mechanism of ET-1 interfere insulin signaling in VSMCs remains unclear. To characterize insulin signaling during modest insulin resistant syndrome, we established and monitored rats by feeding high fructose-diet (HFD) until high blood pressure and modest insulin resistance occurred. To explore the role of ET-1/ETAR during insulin resistance, ETAR expression, ET-1 binding, and insulin signaling were investigated in the HFD-fed rats and cultured A-10 VSMCs. Results showed that high blood pressure, tunica medial wall thickening, plasma ET-1 and insulin, and accompanied with modest insulin resistance without overweight and hyperglycemia occurred in early-stage HFD-fed rats. In the endothelium-denuded aorta from HFD-fed rats, ETAR expression, but not ETBR, and ET-1 binding in aorta were increased. Moreover, decreasing of insulin-induced Akt phosphorylation and increasing of insulin-induced ERK phosphorylation were observed in aorta during modest insulin resistance. Interestingly, in ET-1 pretreated VSMCs, the increment of insulin-induced Akt phosphorylation was decreased whereas the increment of insulin-induced ERK phosphorylation was increased. In addition, insulin potentiated ET-1-induced VSMCs migration and proliferation due to increasing ET-1 binding. ETAR antagonist reversed effects of ET-1 on insulin-induced signaling and VSMCs migration and proliferation. In summary, modest insulin resistance syndrome accompanied with hyperinsulinemia leading to the potentiation on ET-1-induced actions in aortic VSMCs. ET-1 via ETAR pathway suppressed insulin-induced AKT activation, whereas remained insulin-induced ERK activation. ET-1 and insulin synergistically potentiated migration and proliferation mainly through ETAR/ERK dependent pathway, which is dominant in VSMCs during modest insulin resistance syndrome. Therefore, ET-1 and ETAR are potential targets responsible for the observed synergism effect in the hypertensive atherosclerotic process through enhancement of ET-1 binding, ET-1 binding, ETAR expression, and ET-1-induced mitogenic actions in aortic VSMCs.


Assuntos
Aterosclerose/etiologia , Endotelina-1/fisiologia , Hipertensão/etiologia , Resistência à Insulina , Animais , Aterosclerose/fisiopatologia , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Endotelina-1/metabolismo , Hipertensão/fisiopatologia , Insulina/administração & dosagem , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley
16.
J Surg Res ; 194(2): 667-678, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25575733

RESUMO

BACKGROUND: Atherosclerosis is an inflammatory disease with the most common pathologic process leading to cardiovascular diseases. The aim of this study was to evaluate the effect of artemisinin (ART) on the proliferation, migration, and inflammation induced by tumor necrosis factor-α (TNF-α) of rat vascular smooth muscle cells (VSMCs). MATERIALS AND METHODS: Primary rat VSMCs were pretreated with ART and then co-incubated with TNF-α. Cell proliferation was evaluated by MTT assay. Cell migration was assessed by transwell assay. Reactive oxygen species (ROS) production was measured by flow cytometry after staining with dichloro-dihydro-fluorescein diacetate. Inflammation factors of nitric oxide and prostaglandin E2 (PGE2) were measured by responding assay kits. Expression levels of nuclear factor kappa B (NF-κB) subunit NF-κB p65 and the regulator inhibitor of nuclear factor kappa-B kinase-alpha (IκBα) were tested by Western blot, meanwhile, the activation of NF-κB was observed by immunofluorescence assay. RESULTS: The proliferation, migration, and inflammation of VSMCs induced by TNF-α were significantly inhibited by ART treatment in a dose-dependent manner. Treatment with 100 µM ART for 2 h significantly reduced the expression of proliferating cell nuclear antigen and migration-related proteins matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). On the other hand, the same treatment decreased the inflammation factors production of nitric oxide and PGE2. Fluorescence-activated cell sorting analysis revealed that ART suppressed the ROS production induced by TNF-α. Western blot analysis showed that both inflammation mediators inducible nitric oxide synthase and cyclooxygenase and the NF-κB pathway subunit NF-κB p65 were downregulated by ART. CONCLUSIONS: The results suggest that ART can effectively inhibit the proliferation, migration, and inflammation of VSMCs induced by TNF-α through ROS-mediated NF-κB signal pathway.


Assuntos
Anti-Infecciosos/uso terapêutico , Artemisia , Artemisininas/uso terapêutico , Aterosclerose/prevenção & controle , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Anti-Infecciosos/farmacologia , Aorta Torácica/citologia , Artemisininas/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas I-kappa B/metabolismo , Inflamação/tratamento farmacológico , Masculino , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fitoterapia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
17.
Cytokine ; 68(1): 40-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24656930

RESUMO

An anti-inflammatory cytokine, interleukin-10 (IL-10) exerts inhibitory effects on vascular inflammation. Chemokines promote vascular inflammation and play a pathogenic role in the development and maintenance of hypertension. However, chemokine CCL5 has down-regulatory effects on angiotensin II (Ang II)-induced hypertensive mediators. In the present study, IL-10 increased CCL5 expression and attenuated Ang II-induced CCL5 inhibition significantly in vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR), whereas TGF-ß had no effect on CCL5 expression or Ang II-induced CCL5 inhibition. Increased CCL5 expression due to IL-10 was mediated mainly through AT2 R activation. Additionally, IL-10 increased activation of AMP-activated protein kinase (AMPK), which further mediated the up-regulatory effect of IL-10 on CCL5 expression. Attenuation of Ang II-induced CCL5 inhibition by IL-10 was associated with suppression of NF-кB activation, and IL-10 inhibited both Ang II-induced IкB-α and IкB-ß degradation in SHR VSMCs. Moreover, IL-10 partially mediated the inhibitory effects of CCL5 on Ang II-induced 12-lipoxygenase (LO) and endothelin (ET)-1 expression in SHR VSMCs. Taken together, this study provides novel evidence that IL-10 plays an up-regulatory role in the anti-hypertensive activity of CCL5 in SHR VSMCs.


Assuntos
Quimiocina CCL5/metabolismo , Interleucina-10/fisiologia , Regulação para Cima/fisiologia , Adenilato Quinase/metabolismo , Angiotensina II/farmacologia , Animais , Células Cultivadas , Quimiocina CCL5/antagonistas & inibidores , Quimiocina CCL5/genética , Ensaio de Desvio de Mobilidade Eletroforética , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase em Tempo Real
18.
Cytokine ; 67(2): 77-84, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24656927

RESUMO

AMP-activated protein kinase (AMPK) activation plays a central role in cellular metabolic homeostasis. Although AMPK is known for its roles in energy homeostasis, numerous recent studies have suggested broader protective roles in inflammation and hypertension. Chemokine CCL5 has shown down-regulatory effects on angiotensin II (Ang II)-induced hypertensive mediators as well as VSMCs proliferation in spontaneously hypertensive rats (SHR) VSMCs. In the present study, we investigated the relationship between CCL5 and AMPK in the anti-hypertensive effects of CCL5 in SHR VSMCs. CCL5 increased AMPK phosphorylation and attenuated Ang II-induced AMPK inhibition. AMPK activation induced by CCL5 was mediated mainly through the AT2 R pathway. Activation of dimethylarginine dimethylaminohydrolase (DDAH)-1 by CCL5 resulted in AMPK activation as well as attenuation of Ang II-induced AMPK inhibition. In addition, AMPK activation induced by CCL5 was partially responsible for the inhibitory effects of CCL5 on Ang II-induced 12-lipoxygenase (12-LO) and endothelin (ET)-1 expression, and the inhibitory effect of CCL5 on Ang II-induced VSMCs proliferation was also mediated via AMPK activation in SHR VSMCs. In conclusion, CCL5 induces activation of AMPK via DDAH-1 activity in SHR VSMCs, and activation of AMPK is partially responsible for the inhibitory effects of CCL5 on Ang II-induced hypertensive mediators. These results suggest that activation of AMPK by CCL5 potentially expands the anti-hypertensive role of CCL5 in SHR VSMCs.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Quimiocina CCL5/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Amidoidrolases/genética , Amidoidrolases/metabolismo , Angiotensina II/farmacologia , Animais , Western Blotting , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Masculino , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Interferência de RNA , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Front Genet ; 15: 1345459, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38469117

RESUMO

Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.

20.
Biomolecules ; 14(3)2024 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-38540696

RESUMO

Calcification is a process of accumulation of calcium in tissues and deposition of calcium salts by the crystallization of PO43- and ionized calcium (Ca2+). It is a crucial process in the development of bones and teeth. However, pathological calcification can occur in almost any soft tissue of the organism. The better studied is vascular calcification, where calcium salts can accumulate in the intima or medial layer or in aortic valves, and it is associated with higher mortality and cardiovascular events, including myocardial infarction, stroke, aortic and peripheral artery disease (PAD), and diabetes or chronic kidney disease (CKD), among others. The process involves an intricate interplay of different cellular components, endothelial cells (ECs), vascular smooth muscle cells (VSMCs), fibroblasts, and pericytes, concurrent with the activation of several signaling pathways, calcium, Wnt, BMP/Smad, and Notch, and the regulation by different molecular mediators, growth factors (GFs), osteogenic factors and matrix vesicles (MVs). In the present review, we aim to explore the cellular players, molecular pathways, biomarkers, and clinical treatment strategies associated with vascular calcification to provide a current and comprehensive overview of the topic.


Assuntos
Cálcio , Calcificação Vascular , Humanos , Cálcio/metabolismo , Células Endoteliais/metabolismo , Sais , Transdução de Sinais , Calcificação Vascular/metabolismo , Células Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa