Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 411
Filter
1.
Chinese Pharmacological Bulletin ; (12): 62-69, 2024.
Article in Chinese | WPRIM | ID: wpr-1013591

ABSTRACT

Aim To study the effect of menthol on hypobaric hypoxia-induced pulmonary arterial hypertension and explore the underlying mechanism in mice. Methods 10 to 12 weeks old wild type (WT) mice and TRPM8 gene knockout (TRPM8

2.
Journal of Medical Biomechanics ; (6): E156-E163, 2023.
Article in Chinese | WPRIM | ID: wpr-987929

ABSTRACT

Objective To investigate the effect of pathologically elevated-cyclic stretch induced by hypertension on mitochondrial biogenesis of vascular smooth muscle cells (VSMCs), and the role of PGC1α in this process. Methods The Flexcell-5000T stretch loading system in vitro was applied to VSMCs with a frequency of 1. 25 Hz and an amplitude of 5% or 15% to simulate the mechanical environment under normal physiological or hypertensive pathological conditions respectively. Western blotting and qPCR were used to detect the expression of PGC1α, citrate synthase and mitochondrial DNA (mtDNA) copy number in VSMCs under normal physiological or hypertensive pathological conditions. VSMCs were treated with PGC1α specific activator ZLN005 to promote PGC1α expression or specific interfering fragment siRNA to inhibit PGC1α expression in order to detect the effect on citrate synthase and mtDNA copy number. Results Compared with 5% physiological cyclic stretch, 15% pathologically elevated-cyclic stretch significantly suppressed the expression of PGC1α, citrate synthase and mtDNA copy number in VSMCs. Compared with control group, the protein expression of PGC1α was significantly decreased and increased respectively. When VSMCs transfected with PGC1α siRNA or incubated PGC1α activator ZLN005, the expression of citrate synthase and mtDNA copy number were also significantly down regulated and up-regulated in VSMCs accordingly. Under physiological cyclic stretch conditions, the protein level of PGC1α was significantly down-regulated by PGC1α siRNA, which also significantly down-regulated citrate synthase expression and mtDNA copy number. The protein expression of PGC1α was significantly up-regulated by ZLN005, which also enhanced the expression of citrate synthase and mtDNA copy number. Conclusions The pathological cyclic stretch induced by hypertension significantly down-regulated the expression of citrate synthase and mtDNA copy number via suppressing the expression of PGC1α, resulting in mitochondrial dysfunction of VSMCs. PGC1α may be a potential therapeutic target molecule to alleviate the progression of hypertension.

3.
Chinese Journal of Geriatrics ; (12): 720-725, 2023.
Article in Chinese | WPRIM | ID: wpr-993881

ABSTRACT

Objective:To explore the effects of different concentrations of 2-hydroxybenzylamine(2-HOBA)on atherosclerosis and vascular smooth muscle cell senescence and the underlying mechanisms.Methods:Fourteen apolipoprotein E-deficient(ApoE-/-)mice were used to establish an atherosclerosis model and were divided into two groups(n=7)using the random number method: a high-fat diet(HD)group and a high-fat diet plus 2-HOBA(1 mg/ml)(HD+ HOBA)group.Pulse wave velocity was used to assess vascular stiffness and a treadmill was used to assess exercise endurance.Oil Red O staining was used to detect the size and number of atherosclerotic plaques.Masson staining was used to detect the morphology of collagen fibers and elastic fibers in the plaque, the size of the necrotic core area of the plaque, and the thickness of the fibrous cap.Mouse smooth muscle cells were treated with different concentrations of 2-HOBA(100 μmol/L, 250 μmol/L and 500 μmol/L)to establish an H 2O 2-induced senescence model.Senescence-associated β-galactosidase staining was used to detect cell senescence.Real-time quantitative polymerase chain reaction(RT-qPCR)was used to detect the mRNA expression levels of senescence-related secretory phenotype factors, and Western blot was used to detect the expression levels of senescence-related signaling proteins. Results:Compared with the HD group, the HD+ HOBA group showed that the area and number of aortic atherosclerotic plaques were decreased, and the atherosclerotic plaques were stabilized.In addition, compared with the HD group, vascular stiffness in the HD+ 2-HOBA group decreased by 26%(2.59±0.32 mm/ms vs.3.50±0.28 mm/ms), with a statistically significant difference( P<0.01), and exercise endurance increased by 62%[(143.74±24.25)m vs.(233.50±30.21)m, P<0.01], suggesting that 2-HOBA was able to improve aortic vascular stiffness and exercise endurance in mice.2-HOBA ameliorated H 2O 2-induced vascular smooth muscle cell senescence and decreased the mRNA levels of H 2O 2-induced senescence-associated secretory phenotype factors such as interleukin-1β, interleukin-6, tumor necrosis factor-α and monocyte chemoattractant protein-1.Meanwhile, 2-HOBA also inhibited the expression of p53 and p21, the key signaling factors of senescence. Conclusions:2-HOBA suppresses the development and progression of atherosclerosis through inhibiting oxidative stress-related p53/p21 signaling activation and ameliorating vascular smooth muscle cell senescence and the aging-related inflammatory phenotype.

4.
Chinese Journal of Clinical Pharmacology and Therapeutics ; (12): 114-120, 2023.
Article in Chinese | WPRIM | ID: wpr-1014706

ABSTRACT

Pulmonary hypertension (PH) is a rare and severe progressive disease. It results from hypertrophic remodeling of distal pulmonary arterioles that increases pulmonary arterial pressure and pulmonary vascular resistance in the absence of left heart, pulmonary parenchymal, or thromboembolic disease. Hypoxia-inducible factor-1 (HIF-1) regulates a large number of genes related to the occurrence and development of PH, and induces pulmonary angiogenesis, cell proliferation and migration, cellular energy metabolism and utilization. HIF-1 is an important component of the pathogenesis of hypoxic PH and plays an important role in driving the pathological process of pulmonary vascular and right ventricular remodeling. This article systematically elucidated the role and regulation of HIF-1 in hypoxic PH and its potential in targeted therapy of PH.

5.
Chinese Pharmacological Bulletin ; (12): 532-536, 2023.
Article in Chinese | WPRIM | ID: wpr-1013826

ABSTRACT

Aim To investigate the effect of terpinen-4-ol (T40) on inflammatory injury of vascular smooth muscle cells (VSMCs) induced by high glucose based on the improvement of autophagic flow disorder and involved molecular signals. Methods The scratch test was used to analyze the migration ability of VSMCs, the levels of IL-1β and IL-6 in cell culture supernatant were measured by ELISA, the expression levels of inflammation-related proteins NF-κb p65, p-NF-κb p65, IL-1β, IL-18 and autophagy-related proteins p62, LC3-HYLC3-I, Beclinl, p-Beclinl were de-tected by Western blot. Results T40 inhibited migration of VSMCs induced by high glucose, reduced the secretion and release of pro-inflammatory factors IL-1β and IL-6, inhibited the expression of p-NF-κb p65/ NF-κb p65, IL-1β, IL-18, downregulated the expression of p62, LC3-TJ/LC3- I and p-Beclinl at same time. After interfering the autophagic flux of VSMCs with autophagy inhibitor chloroquine (CQ) , T40 pre-treatment significantly inhibited the protein expression levels of the above inflammatory factors and autophagy-related signals which mediated by CQ. Conclusion T40 inhibits the inflammatory injury of VSMCs induced by high glucose through improving the autophagic flow disorder.

6.
Chinese Pharmacological Bulletin ; (12): 1829-1835, 2023.
Article in Chinese | WPRIM | ID: wpr-1013695

ABSTRACT

Aim To explore the effect of LINC00707 on the proliferation, migration and inflammatory factors of human vascular smooth muscle cells induced by oixidized low-density lipoprotein (ox-LDL) and its possible mechanism. Methods ox-LDL was used to induce human vascular smooth muscle cells (HVSMCs) to establish an atherosclerotic cell model. si-NC, siLINC00707, miR-NC, miR-30c-5p mimic were trans-fected into HVSMCs, and then 100 mg • L~ ox-LDL was added to the cells. si-LINC00707 and anti-miRNC, or si-LINC00707 and miR-30c-5p Inhibitor were co-transfected into HVSMCs and then treated with 100 mg • L ox-LDL. qRT-PCR was used to detect the expression of LINC00707 and miR-30c-5p. CCK-8 and Transwell test were used to detect cell proliferation and migration. ELISA was used to detect the levels of IL-6, TNF-a, and IL-10. The dual-luciferase reporter experiment was used to detect the targeting relationship between LINC00707 and miR-30c-5p. Western blot was used to detect the protein expression of E-cadherin and N-cadherin. Results The expression of LINC00707 in HVSMCs induced by ox-LDL increased (P <0. 05), while the expression of miR-30c-5p decreased (P < 0. 05). After transfection with siLINC00707 or miR-30c-5p mimic, cell viability, the protein level of N-cadherin, the levels of IL-6 and TNF-a decreased (P < 0. 05), and the number of im grating cells decreased (P<0. 05), while the protein level of E-coadherin and the level of IL-10 increased (P <0. 05). LINC00707 could target miR-30c-5p. After co-transfection with si-LINC00707 and miR-30c-5p inhibitor, cell survival rate, the protein level of N-cadherin, the levels of IL-6 and TNF-α increased (P < 0.05), and the number of migrating cells increased (P <0. 05), while the protein level of E-cadherin and the level of IL-10 decreased (P < 0. 05). Conclusion Down-regulation of the expression of LINC00707 could inhibit the proliferation, migration and inflammation of human vascular smooth muscle cells induced by ox-LDL by promoting the expression of miR-30c-5p.

7.
Acta Pharmaceutica Sinica B ; (6): 2369-2382, 2023.
Article in English | WPRIM | ID: wpr-982871

ABSTRACT

Pulmonary hypertension (PH) is an insidious pulmonary vasculopathy with high mortality and morbidity and its underlying pathogenesis is still poorly delineated. The hyperproliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs) contributes to pulmonary vascular remodeling in pulmonary hypertension, which is closely linked to the downregulation of fork-head box transcriptional factor O1 (FoxO1) and apoptotic protein caspase 3 (Cas-3). Here, PA-targeted co-delivery of a FoxO1 stimulus (paclitaxel, PTX) and Cas-3 was exploited to alleviate monocrotaline-induced pulmonary hypertension. The co-delivery system is prepared by loading the active protein on paclitaxel-crystal nanoparticles, followed by a glucuronic acid coating to target the glucose transporter-1 on the PASMCs. The co-loaded system (170 nm) circulates in the blood over time, accumulates in the lung, effectively targets the PAs, and profoundly regresses the remodeling of pulmonary arteries and improves hemodynamics, leading to a decrease in pulmonary arterial pressure and Fulton's index. Our mechanistic studies suggest that the targeted co-delivery system alleviates experimental pulmonary hypertension primarily via the regression of PASMC proliferation by inhibiting cell cycle progression and promoting apoptosis. Taken together, this targeted co-delivery approach offers a promising avenue to target PAs and cure the intractable vasculopathy in pulmonary hypertension.

8.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 83-92, 2023.
Article in Chinese | WPRIM | ID: wpr-969602

ABSTRACT

ObjectiveTo investigate whether the effects of paeonol (Pae) on angiotensin Ⅱ (AngⅡ)-induced senescence in vascular smooth muscle cells (VSMCs) were related to angiotensinogen of silencing regulatory information factor 6 (SIRT6)/adenosine diphosphate ribose polymerase 1 (PARP1) signaling pathway in VSMCs. MethodThe model of VSMC-stress aging induced by AngⅡ (100 nmol·L-1) was established. The rats were divided into normal group, model group, low, medium, and high-concentration Pae groups (30, 60, 120 μmol·L-1). The positive rate of cell senescence was detected by SA-β-Gal staining, the ability of cell proliferation was detected by the cell counting kit-8 (CCK-8) method, the expression of SIRT6, PARP1, p16, p21, p53, proliferating cell nuclear antigen (PCNA), deoxyribonucleic acid (DNA)-damaged protein γ-H2AX was detected by Western blot, and VSMC proliferation was detected by EdU staining. The silenced VSMCs were prepared by siRNA-SIRT6 transfection, and the protein expressions of SIRT6, PARP1, p16, and γ-H2AX in VSMCs silenced by SIRT6 were observed. ResultThe results of SA-β-Gal staining showed that the senescence positive rate of SA-β-Gal staining in the model group was higher than that in the normal group (P<0.01), and the positive rate of SA-β-Gal staining in the Pae group was significantly lower than that in the model group (P<0.05, P<0.01). The results of Western blot showed that as compared with the normal group, the expression of PCNA, SIRT6, and PARP1 in the model group was down-regulated, and the expression of aging-related proteins p16, p21, p53, and γ-H2AX was up-regulated in the model group (P<0.05, P<0.01). Compared with the model group, Pae promoted the protein expression of PCNA, SIRT6, and PARP1 and inhibited the protein expression of p16, p21, p53, and γ-H2AX in a dose-dependent manner (P<0.05, P<0.01). The results of EdU staining showed that the number of EdU positive cells in the model group was lower than that in the normal group (P<0.01), and the number of EdU positive cells in Pae groups was significantly higher than that in the model group (P<0.05, P<0.01). After SIRT6 silencing, the effects of Pae on promoting SIRT6 and PARP1 and inhibiting P16 were reversed (P<0.05, P<0.01). In addition, the addition of SIRT6 inhibitor (IN-1) promoted the occurrence of cell senescence induced by AngⅡ (P<0.05, P<0.01). ConclusionPae can effectively inhibit the aging of VSMCs, and its mechanism may be related to the regulation of SIRT6/PARP1 signal pathway.

9.
China Journal of Chinese Materia Medica ; (24): 2530-2537, 2023.
Article in Chinese | WPRIM | ID: wpr-981329

ABSTRACT

This study aimed to observe the effect of terpinen-4-ol(T4O) on the proliferation of vascular smooth muscle cells(VSMCs) exposed to high glucose(HG) and reveal the mechanism via the Krüppel-like factor 4(KLF4)/nuclear factor kappaB(NF-κB) signaling pathway. The VSMCs were first incubated with T4O for 2 h and then cultured with HG for 48 h to establish the model of inflammatory injury. The proliferation, cell cycle, and migration rate of VSMCs were examined by MTT method, flow cytometry, and wound healing assay, respectively. The content of inflammatory cytokines including interleukin(IL)-6 and tumor necrosis factor-alpha(TNF-α) in the supernatant of VSMCs was measured by enzyme-linked immunosorbent assay(ELISA). Western blot was employed to determine the protein levels of proliferating cell nuclear antigen(PCNA), Cyclin D1, KLF4, NF-κB p-p65/NF-κB p65, IL-1β, and IL-18. The KLF4 expression in VSMCs was silenced by the siRNA technology, and then the effects of T4O on the cell cycle and protein expression of the HG-induced VSMCs were observed. The results showed that different doses of T4O inhibited the HG-induced proliferation and migration of VSMCs, increased the percentage of cells in G_1 phase, and decreased the percentage of cells in S phase, and down-regulated the protein levels of PCNA and Cyclin D1. In addition, T4O reduced the HG-induced secretion and release of the inflammatory cytokines IL-6 and TNF-α and down-regulated the expression of KLF4, NF-κB p-p65/NF-κB p65, IL-1β, and IL-18. Compared with si-NC+HG, siKLF4+HG increased the percentage of cells in G_1 phase, decreased the percentage of cells in S phase, down-regulated the expression of PCNA, Cyclin D1, and KLF4, and inhibited the activation of NF-κB signaling pathway. Notably, the combination of silencing KLF4 with T4O treatment further promoted the changes in the above indicators. The results indicate that T4O may inhibit the HG-induced proliferation and migration of VSMCs by down-regulating the level of KLF4 and inhibiting the activation of NF-κB signaling pathway.


Subject(s)
NF-kappa B/metabolism , Interleukin-18/metabolism , Proliferating Cell Nuclear Antigen/genetics , Cyclin D1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Muscle, Smooth, Vascular , Cell Proliferation , Signal Transduction , Cytokines/metabolism , Glucose/metabolism
10.
China Pharmacy ; (12): 1066-1070, 2023.
Article in Chinese | WPRIM | ID: wpr-972948

ABSTRACT

OBJECTIVE To study the inhibitory effect mechanism of rhynchophylline solid lipid nanoparticles (Rhy-SLN) on the proliferation of airway smooth muscle cells (ASMCs) in asthmatic model mice. METHODS Asthma model was prepared by ovalbumin+calmogastrin sensitization. The primary isolation and culture of ASMCs were performed, and morphological observation and identification were also conducted [when α -smooth muscle actin (α -SMA) appeared red and Desmin appeared green in ASMCs, indicating successful cultivation of ASMCs]. The cells were divided into blank group (ASMCs of normal mice), model group (ASMCs of asthma model mice), Rhy-SLN group (ASMCs of asthma model mice), recombinant suppressors of cytokine signaling 1 (SOCS1) overexpression group (ASMCs of asthma model mice transfected with SOCS1 vector), SOCS1-RNAi group (ASMCs of asthma model mice transfected with SOCS1-RNAi vector) and SB203580 group [p38 mitogen-activated protein kinase (p38 MAPK) inhibitor, ASMCs of asthma model mice]. The cells of each group were added into the corresponding culture medium containing drug (10 μmol/L) or not containing drug for 24 hours. MTT method was used to detect the proliferation of ASMCs in asthmatic mice; Western blot assay was used to detect the protein expressions of α-SMA, interleukin-1β (IL-1β), SOCS1, p38 MAPK and phosphorylated p38 MAPK (p-p38 MAPK) in ASMCs. RESULTS The primary ASMCs of mice varied in shape and size, presenting irregular, spindle and triangular shapes;α-SMA appeared red and Desmin appeared green, indicating successful cultivation of ASMCs. Compared with model group, ASMCs absorbance values and protein expressions of α -SMA, p38 MAPK, and p-p38 MAPK were reduced significantly in Rhy- SLN group, SOCS1 overexpression group and SB203580 E-mail:wangmeng106@163.com group, while protein expression of SOCS1 (except for group) was increased significantly (P<0.05); protein expressions of IL-1β was reduced significantly in ASMCs (P< 0.05). ASMCs absorbance values and protein expressions of α-SMA, SOCS1, p38 MAPK and p-p38 MAPK were increased significantly in SOCS1-RNAi group (P<0.05). CONCLUSIONS Rhy-SLN can inhibit the proliferation of ASMCs, the mechanism of which may be associated with overexpression of SOCS1 and inhibiting the protein expressions of IL-1β and p38 MAPK.

11.
Journal of Zhejiang University. Medical sciences ; (6): 750-757, 2023.
Article in English | WPRIM | ID: wpr-971092

ABSTRACT

Phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs) is a key factor in pulmonary vascular remodeling. Inhibiting or reversing phenotypic transformation can inhibit pulmonary vascular remodeling and control the progression of hypoxic pulmonary hypertension. Recent studies have shown that hypoxia causes intracellular peroxide metabolism to induce oxidative stress, induces multi-pathway signal transduction, including those related to autophagy, endoplasmic reticulum stress and mitochondrial dysfunction, and also induces non-coding RNA regulation of cell marker protein expression, resulting in PASMCs phenotypic transformation. This article reviews recent research progress on mechanisms of hypoxia-induced phenotypic transformation of PASMCs, which may be helpful for finding targets to inhibit phenotypic transformation and to improve pulmonary vascular remodeling diseases such as hypoxia-induced pulmonary hypertension.


Subject(s)
Humans , Pulmonary Artery , Hypertension, Pulmonary , Vascular Remodeling/genetics , Hypoxia/genetics , Myocytes, Smooth Muscle , Cell Proliferation/physiology , Cells, Cultured , Cell Hypoxia/genetics
12.
China Journal of Chinese Materia Medica ; (24): 1446-1454, 2023.
Article in Chinese | WPRIM | ID: wpr-970616

ABSTRACT

Tetramethylpyrazine is the main component of Ligusticum chuanxiong. Studies have found that tetramethylpyrazine has a good protective effect against cardiovascular diseases. In the heart, tetramethylpyrazine can reduce myocardial ischemia/reperfusion injury by inhibiting oxidative stress, regulating autophagy, and inhibiting cardiomyocyte apoptosis. Tetramethylpyrazine can also reduce the damage of cardiomyocytes caused by inflammation, relieve the fibrosis and hypertrophy of cardiomyocytes in infarcted myocardium, and inhibit the expansion of the cardiac cavity after myocardial infarction. In addition, tetramethylpyrazine also has a protective effect on the improvement of familial dilated cardiomyopathy. Besides, the mechanisms of tetramethylpyrazine on blood vessels are more abundant. It can inhibit endothelial cell apoptosis by reducing oxidative stress, maintain vascular endothelial function and homeostasis by inhibiting inflammation and glycocalyx degradation, and protect vascular endothelial cells by reducing iron overload. Tetramethylpyrazine also has a certain inhibitory effect on thrombosis. It can play an anti-thrombotic effect by reducing inflammatory factors and adhesion molecules, inhibiting platelet aggregation, and suppressing the expression of fibrinogen and von Willebrand factor. In addition, tetramethylpyrazine can also reduce the level of blood lipid in apolipoprotein E-deficient mice, inhibit the subcutaneous deposition of lipids, inhibit the transformation of macrophages into foam cells, and inhibit the proliferation and migration of vascular smooth muscle cells, thereby reducing the formation of atherosclerotic plaque. In combination with network pharmacology, the protective mechanism of tetramethylpyrazine on the cardiovascular system may be mainly achieved through the regulation of phosphatidylinositol 3 kinase/protein kinase B(PI3K/Akt), hypoxia-inducible factor 1(HIF-1), and mitogen-activated protein kinase(MAPK) pathways. Tetramethylpyrazine hydrochloride and sodium chloride injection has been approved for clinical application, but some adverse reactions have been found in clinical application, which need to be paid attention to.


Subject(s)
Mice , Animals , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Myocardial Infarction , Myocardium/metabolism , Myocytes, Cardiac , Thrombosis , Inflammation , Apoptosis
13.
Rev. bras. cir. cardiovasc ; 38(6): e20220260, 2023. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1514975

ABSTRACT

ABSTRACT Introduction: Thoracic aortic aneurysm is a potentially fatal disease with a strong genetic contribution. The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the formation of this aneurysm. Although previous studies suggested that long non-coding ribonucleic acid (RNA) hypoxia inducible factor 1 α-antisense RNA 1 (HIF1A-AS1) exerted a vital role in the progression and pathogenesis of thoracic aortic aneurysm, we managed to find a new regulatory mechanism of HIF1A-AS1 in VSMCs via transcriptomics. Methods: Cell viability was detected by the cell counting kit-8 assay. Cell apoptosis was assessed by Annexin V-fluorescein isothiocyanate/propidium iodide double staining. Transwell migration assay and wound healing assay were performed to check the migration ability of HIF1A-AS1 on VSMCs. The NextSeq XTen system (Illumina) was used to collect RNA sequencing data. Lastly, reverse transcription-quantitative polymerase chain reaction confirmed the veracity and reliability of RNA-sequencing results. Results: We observed that overexpressing HIF1A-AS1 successfully promoted apoptosis, significantly altered cell cycle distribution, and greatly attenuated migration in VSMCs, further highlighting the robust promoting effects of HIF1A-AS1 to thoracic aortic aneurysm. Moreover, transcriptomics was implemented to uncover its underlying mechanism. A total of 175 differently expressed genes were identified, with some of them enriched in apoptosis, migration, and cell cycle-related pathways. Intriguingly, some differently expressed genes were noted in vascular development or coagulation function pathways. Conclusion: We suggest that HIF1A-AS1 mediated the progression of thoracic aortic aneurysm by not only regulating the function of VSMCs, but also altering vascular development or coagulation function.

14.
An. bras. dermatol ; 97(6): 716-728, Nov.-Dec. 2022. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1403177

ABSTRACT

Abstract Background: To evaluate the effect of T-helper 17 (Th17) cells and Th9 cells on the activation of dermal vascular smooth muscle cells (DVSMCs) in systemic scleroderma (SSc) and regulation of tanshinone IIA. Methods: The expression of interleukin 17 receptor (IL-17R) and interleukin 9 receptor (IL-9R) in the skin of SSc patients was assessed by immunofluorescence. The expression of IL-9 and IL-9R mRNA in peripheral blood mononuclear cells (PBMCs) of SSc patients were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The proportion of Th9 cells in PBMCs of SSc patients was sorted by flow cytometry. The effect of IL-9 on the differentiation of Th17 and IL-17 on that of Th9 was detected by flow cytometry. The proportion of Th9 and Th17 cells in SSc patients was detected by flow cytometry. The level of collagen I, III, α-SMA, IL-9R, IL-17R, JNK, P38, and ERK were analyzed using western blot (WB). Results: Th9 cells were highly expressed in SSc. IL-9 stimulated the differentiation of immature T cells into Th17 cells. IL-17 induced the differentiation of immature T cells intoTh9 cells. Tanshinone IIA inhibited the differentiation of immature T lymphocytes into Th17 and Th9. WB showed that the combined action of IL-17 and IL-9 upregulated the inflammation and proliferation of DVSMCs. Anti-IL17, anti-IL9, and tanshinone IIA inhibited the functional activation of DVSMCs. Study limitations: For Th17, Th9 and vascular smooth muscle cells, the study on the signal pathway of their interaction is not thorough enough. More detailed studies are needed to explore the mechanism of cell-cell interaction. Conclusions: The current results suggested that Th17 and Th9 cells induced the activation of DVSMCs in SSc through crosstalk in vitro, and tanshinone IIA inhibited the process.

15.
Int. j. morphol ; 40(3): 760-767, jun. 2022. ilus
Article in English | LILACS | ID: biblio-1385669

ABSTRACT

SUMMARY: Atherosclerosis is a complex disease whose pathogenesis includes endothelial activation, accumulation of lipids in the subendothelium, formation of foam cells, fat bands and formation of atherosclerotic plaque. These complex mechanisms involve different cell populations in the intimate sub-endothelium, and the S-100 protein family plays a role in a number of extracellular and intracellular processes during the development of atherosclerotic lesions. The aim of this study was to determine the phenotypic characteristics of smooth muscle cells and the consequent expression of S100 protein in atherosclerotic altered coronary arteries in advanced stages of atherosclerosis. 19 samples of right atherosclerotic coronary arteries in stages of fibro atheroma (type V lesion) and complicated lesions (type VI lesion) have been analyzed. According to the standard protocol, the following primary antibodies have been used in the immunohistochemical analysis: a-smooth muscle actin (α-SMA), vimentin and S-100 protein. All analyzed samples have been in advanced stages of atherosclerosis, fibro atheroma (stage V lesions) and complicated lesions (type VI lesions). Most of them have had the structure of a complicated lesion with atheroma or fibro atheroma as a basis, subsequently complicated by disruption (subtype VI a), hemorrhage (subtype VI b) or thrombosis (subtype VI c), as well as by the presence of several complications on the same sample. Marked hypocellularity is present in the subendothelium of plaques. Cell population at plaque margins is characterized by immunoreactivity to α-SMA, vimentin, and S100 protein. Some of these cells accumulate lipids and look like foam cells. In the cell population at the margins of the plaques, smooth muscle cells of the synthetic phenotype are present, some of which accumulate lipids and demonstrate S100 immunoreactivity. Summarizing numerous literature data and our results, we could assume that smooth muscle cells, due to their synthetic and proliferative activity in the earlier stages of pathogenesis, as well as the consequent expression of S100 protein, could accumulate lipids in the earlier stages of atherosclerosis which, in advanced stages analyzed in this study, result in immunoreactivity of foam cells of smooth muscle origin to S100 protein.


RESUMEN: La aterosclerosis es una enfermedad compleja cuya patogenia incluye activación endotelial, acumulación de lípidos en el subendotelio, formación de células espumosas, bandas grasas y formación de placa aterosclerótica. Estos complejos mecanismos involucran diferentes poblaciones celulares en el subendotelio íntimo, y la familia de proteínas S-100 juega un papel en varios procesos extracelulares e intracelulares durante el desarrollo de lesiones ateroscleróticas. El objetivo de este estudio fue determinar las características fenotípicas de las células de músculo liso y la consecuente expresión de la proteína S100 en arterias coronarias alteradas ateroscleróticas en estadios avanzados de aterosclerosis. Se analizaron 19 muestras de arterias coronarias ateroscleróticas derechas en estadios de fibroateroma (lesión tipo V) y lesiones complicadas (lesión tipo VI). Según el protocolo estándar, en el análisis inmunohistoquímico se utilizaron los siguientes anticuerpos primarios: α-actina de músculo liso (α-SMA), vimentina y proteína S-100. Todas las muestras analizadas han estado en estadios avanzados de aterosclerosis, fibroateroma (lesiones estadio V) y lesiones complicadas (lesiones tipo VI). La mayoría de ellos han tenido la estructura de una lesión complicada con ateroma o fibroateroma como base, complicada posteriormente por disrupción (subtipo VI a), hemorragia (subtipo VI b) o trombosis (subtipo VI c), así como por la presencia de varias complicaciones en la misma muestra. La hipocelularidad marcada estaba presente en el subendotelio de las placas. La población celular en los márgenes de la placa se caracterizaba por inmunorreactividad a α-SMA, vimentina y proteína S100. Algunas de estas células acumulan lípidos y parecen células espumosas. En la población celular en los márgenes de las placas, estaban presentes las células de músculo liso de fenotipo sintético, algunas de las cuales acumulaban lípidos y mostraban inmunorreactividad S100. Resumiendo numerosos datos de la literatura y nuestros resultados, podríamos suponer que las células del músculo liso, debido a su actividad sintética y proliferativa en las primeras etapas de la patogénesis, así como la consecuente expresión de la proteína S100, podrían acumular lípidos en las primeras etapas de la aterosclerosis que, en estadios avanzados analizados en este estudio, dan como resultado inmunorreactividad de células espumosas de origen muscular liso a la proteína S100.


Subject(s)
Humans , Coronary Artery Disease/metabolism , S100 Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype
16.
Chinese Journal of Endocrinology and Metabolism ; (12): 1075-1080, 2022.
Article in Chinese | WPRIM | ID: wpr-994286

ABSTRACT

Objective:To investigate the role of stress-inducible protein Sestrin2 (Sesn2) in the improvement of insulin resistance in rat L6 skeletal muscle cells treated with liraglutide.Methods:The establishment of insulin resistance model of rat L6 skeletal muscle cells was induced by palmitate. The experimental cells were divided into control group(Con group), palmitate 0.6 mmol/L treatment group(PA group), palmitate 0.6 mmol/L+ liraglutide 10 nmol/L treatment group(PA+ Lir10 group), palmitate 0.6 mmol/L+ liraglutide 100 nmol/L treatment group(PA+ Lir100 group), and palmitate 0.6 mmol/L+ liraglutide 1 000 nmol/L treatment group(PA+ Lir1000 group). The cell counting kit 8(CCK8) method was used to detect the cell activity in each group. Western blotting was used to detect the expression levels of glucose transporter 4(GLUT4), protein kinase B(Akt), phosphorylated protein kinase B(p-Akt), and Sesn2 protein in L6 cells. L6 cells were transfected with siRNA to inhibit the expression of Sesn2. The cells were treated with palmitate and liraglutide. Western blotting was used to detect the expression levels of Sesn2, Akt, p-Akt, and GLUT4 protein in L6 cells.Results:Compared with Con group, the cell survival rate, p-Akt/Akt ratio, Sesn2, and GLUT4 protein expression in PA group decreased significantly( P<0.05). After liraglutide intervention, the cell activity, p-Akt/Akt ratio, Sesn2, and GLUT4 protein expression of PA+ Lir100 and PA+ Lir1000 groups was increased( P<0.05). After inhibiting the expression of Sesn2, p-Akt/Akt ratio and GLUT4 protein in transfected si-Sesn2 and treated with 0.6 mmol/L palmitate group(PA+ si-Sesn2 group) and transfected si-Sesn2 and treated with 0.6 mmol/L palmitate+ liraglutide 100 nmol/L group (Lir100+ PA+ si-Sesn2 group) were significantly lower than those in transfection negative group (si-Con group; P<0.05). Even after liraglutide intervention, compared with PA+ si-Sesn2 group, p-Akt/Akt ratio and GLUT4 protein expression level were not significantly increased in Lir100+ PA+ si-Sesn2 group ( P>0.05). Conclusions:Palmitate could induce the decrease of p-Akt/Akt ratio and GLUT4 protein expression in L6 cells. Liraglutide upregulates the expression of Sesn2, which leads to the increase of p-Akt/Akt ratio and GLUT4 protein expression and contributes to the improvement of insulin resistance.

17.
Chinese Journal of Gastroenterology ; (12): 429-433, 2022.
Article in Chinese | WPRIM | ID: wpr-1016100

ABSTRACT

Recent studies have found a special class of macrophages, muscularis macrophages (MMs), in the gastrointestinal tract, which interacts with enteric neuron (EN), interstitial cells of Cajal (ICC), and smooth muscle cells (SMC) to maintain normal intestinal motility. MMs can undergo phenotypic and other changes under altered intestinal microbiota, inflammation, or stress, and act on EN, ICC, or SMC through multiple mechanisms, ultimately affecting gastrointestinal motility. This article reviewed the mechanism of gastrointestinal MMs regulating gastrointestinal motility and role in gastrointestinal motility disorder diseases.

18.
Chinese Pharmacological Bulletin ; (12): 261-267, 2022.
Article in Chinese | WPRIM | ID: wpr-1014154

ABSTRACT

Aim To evaluate the effects of Rutaecarpine(Rut)on the expression of SIRT1 and the senescence of vascular smooth muscle cells(VSMCs)induced by angiotensin Ⅱ.Methods VSMC senescencewas induced by exposure to AngⅡ(1 μmol·L-1)for 72 h.VSMCs were treated with different concentrations of Rut(0.3, 1, 3 μmol·L-1).TRPV1 competitive antagonist CAPZ(10 μmol·L-1)and AMPK inhibitor Compound C(1 μmol·L-1)were used to explore whether TRPV1/AMPK mediated the protective effect of Rut.The quantity of senescent cells were determined by senescence-associated SA-β-Gal staining, and the intracellular ROS level was measured by(DCFH-DA)fluorescent probe.The migration ability of VSMCs was evaluated by Wound-healing assay combined with Transwell assay.The protein level of longevity protein SIRT1 and senescence-related proteins p53, p21 and AMPK phosphorylation level were detected by Western blot.Results Rut significantly inhibited Ang Ⅱ-induced VSMC senescence and ROS production and prevented VSMCs migration.Preprocessing of TRPV1 antagonist CAPZ could abolish the protective effect of Rut.Ang Ⅱ inhibited the expression of longevity protein SIRT1.Rut recovered SIRT1 expression in a dose-dependent manner, while prevented the up-regulation of senescence-related proteins p53 and p21.Ang Ⅱ inhibited AMPK phosphorylation, pre-treatment with Rut restored AMPK phosphorylation level.CAPZ and Compound C eliminated the up-regulating function of Rut on SIRT1 expression.Conclusions Rut up-regulates the expression of SIRT1 and prevents the senescence and migration of VSMCs induced by Angiotensin Ⅱ, which is related to activation of the TRPV1/AMPK signaling pathway.

19.
Chinese Pharmacological Bulletin ; (12): 506-511, 2022.
Article in Chinese | WPRIM | ID: wpr-1014110

ABSTRACT

Pulmonary hypertension ( PH) is occult, with no distinctive clinical manifestations and poor prognosis.Pulmonary vascular remodelling is an important pathological feature in which pulmonary artery smooth muscle cell ( PASMCs) pheno- typic switching plays a crucial role.MicroRNA (miRNA) is a class of evolutionary highly conserved single-stranded small non-coding RNA.Recently, an increasing number of scholars have found that miRNA can play an important role in the occurrence and development of PH by regulating the phenotypic switching of PASMCs, which is expected to be a potential target for the prevention and treatment of PH.It has been found that miR NA such as miR-221 , miR-24, miR-15b, miR-96, miR-23a.miR-9, miR-214, miR-20a can promote the phenotypic switching of PASMCs, while miRNA such as miR-21, miR-132, miR-182, miR-449, miR-206 .miR-124, miR-30c, miR-140.miR-17-92 cluster can inhibit it.This article aims to review the research progress on miRNA that mediates PASMCs phenotypic switching in PH from both growth factor-related miRNA and hy- poxia-related miRNA.

20.
Chinese Pharmacological Bulletin ; (12): 492-496, 2022.
Article in Chinese | WPRIM | ID: wpr-1014107

ABSTRACT

Chronic hypoxic lung diseases are major causes of disability and mortality worldwide, which are typically aggravated by hypoxic pulmonary hypertension.The pathogenesis of hypoxic pulmonary hypertension is complex, and its mechanism has not been fully elucidated.The previous studies have shown abnormally elevated levels of free Ca + in the cytoplasm of pulmonary artery smooth muscle cells to be the predominant drivers of pulmonary hypertension , causing continuous contraction and remodeling of the pulmonary vessels.This article briefly summarizes the mechanism of hypoxia-induced imbalance in calcium homeostasis in pulmonary artery smooth muscle cells, together with its related drug research, based on the existing literature.Hypoxia induces an imbalance in calcium homeostasis in pulmonary artery smooth muscle cells by regulating hypoxia-inducible factor-1, K+ , store-operated calcium channel, receptor-operated calcium channel, the Ca +-sensing myosin contractile mechanism by binding to calmodulin, leading to pulmonary vasoconstriction.Ca + can also activate PKC/ MAPKs and PI3K/Akt/mTOR pathways, leading to pulmonary vascular remodeling.

SELECTION OF CITATIONS
SEARCH DETAIL