Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
Int J Cardiol Heart Vasc ; 54: 101500, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39280692

ABSTRACT

Abdominal aortic aneurysms (AAAs) are characterized by permanent dilatation of the abdominal aorta, which is accompanied by inflammation, degradation of the extracellular matrix (ECM) and disruption of vascular smooth muscle cell (VSMC) homeostasis. Endoplasmic reticulum (ER) stress is involved in the regulation of inflammation, oxidative stress and VSMC apoptosis, all of which are critical factors in AAA development. Although several studies have revealed the occurrence of ER stress in AAA development, the specific biological functions of ER stress in AAA development remain largely unknown. Given that targeting ER stress is a promising strategy for treating AAAs, further investigation of the physiological and pathological roles of ER stress in AAA development is warranted.

2.
Aging (Albany NY) ; 16(11): 9369-9385, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38809515

ABSTRACT

Vascular aging exacerbates diabetes-associated vascular damage, a major cause of microvascular and macrovascular complications. This study aimed to elucidate key genes and pathways underlying vascular aging in diabetes using integrated bioinformatics and machine learning approaches. Gene expression datasets related to vascular smooth muscle cell (VSMC) senescence and diabetic vascular aging were analyzed. Differential expression analysis identified 428 genes associated with VSMC senescence. Functional enrichment revealed their involvement in cellular senescence, ECM-receptor interaction, PI3K-Akt and AGE-RAGE signaling pathways. Further analysis of diabetic vascular aging datasets revealed 52 differentially expressed genes, enriched in AMPK signaling, AGE-RAGE signaling, cellular senescence, and VEGF signaling pathways. Machine learning algorithms, including LASSO regression and SVM-RFE, pinpointed six key genes: TFB1M, FOXRED2, LY75, DALRD3, PI4K2B, and NDOR1. Immune cell infiltration analysis demonstrated correlations between diabetic vascular aging, the identified key genes, and infiltration levels of plasma cells, M1 macrophages, CD8+ T cells, eosinophils, and regulatory T cells. In conclusion, this study identified six pivotal genes (TFB1M, FOXRED2, LY75, DALRD3, PI4K2B, and NDOR1) closely associated with diabetic vascular aging through integrative bioinformatics and machine learning approaches. These genes are linked to alterations in the immune microenvironment during diabetic vascular aging. This study provides a reference and basis for molecular mechanism research, biomarker mining, and diagnosis and treatment evaluation of diabetes-related vascular aging.


Subject(s)
Aging , Computational Biology , Machine Learning , Humans , Aging/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Signal Transduction/genetics , Diabetes Mellitus/genetics , Cellular Senescence/genetics
3.
Cardiovasc Diagn Ther ; 14(1): 72-83, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38434568

ABSTRACT

Background: In recent years, a mass of studies have shown that pyroptosis plays an important role in the proliferation of vascular smooth muscle cells (VSMCs). We investigated whether angiotensin II (Ang II) induces the pyroptosis of rat aortic VSMCs and the role of NOD-like receptor family pyrin domain containing 3 (NLRP3) in this process. Additionally, we explored the effect and related mechanism of recombinant tissue factor pathway inhibitor (rTFPI) in Ang II-induced VSMC pyroptosis. Methods: Cultured VSMCs were divided into five groups: control group, Ang II group (1×10-5 mol/L), MCC950 group (NLRP3 inhibitor, 15 nmol/L), Ang II + MCC950 group and Ang II + rTFPI (50 µg/L) group. Cell viability was measured by cell counting kit-8 (CCK8) assays and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays. Propidium iodide (PI) staining and immunofluorescence were performed to determine the pyroptosis of VSMCs. Changes in VSMC ultrastructure were evaluated through transmission electron microscopy. The expression levels of NLRP3, pro-caspase-1, gasdermin D-N (GSDMD-N), and interleukin-1ß (IL-1ß) were determined by western blot analysis. Results: The cell viability, the positive rate of PI staining, and the expression level of GSDMD detected by immunofluorescence in the Ang II group were higher than that in the control group, whereas they all decreased in Ang II + MCC950 group and Ang II + rTFPI group compared with Ang II group (P<0.05). Electron microscopy analysis revealed less extracellular matrix, increased myofilaments, and decreased endoplasmic reticulum, Golgi complex, and mitochondria in Ang II + rTFPI-treated VSMCs than in Ang II-treated VSMCs. The protein expression levels of the pyroptosis-related molecules NLRP3, pro-caspase-1, GSDMD-N, and IL-1ß in Ang II group showed an increasing trend compared with those in control group (P<0.05); however, these expression levels in Ang II + MCC950 and Ang II + rTFPI groups were significantly lower than those in Ang II group (P<0.05). Conclusions: Ang II may induce pyroptosis in VSMCs by activating NLRP3. rTFPI can inhibit Ang II-induced VSMC pyroptosis. Furthermore, rTFPI might exert this effect by inhibiting the NLRP3 pathway and therefore play an important role in the treatment of vascular remodeling induced by hypertension.

4.
Front Cardiovasc Med ; 10: 1172080, 2023.
Article in English | MEDLINE | ID: mdl-37288252

ABSTRACT

There is gaining popularity in the use of single-cell technology and analysis in studying the pathogenesis of abdominal aortic aneurysm (AAA). As there are no current pharmacologic therapies for impeding aneurysm growth or preventing AAA rupture, identifying key pathways involved in AAA formation is critical for the development of future therapies. Single-cell RNA sequencing (scRNA-seq) technology provides an unbiased and global view of transcriptomic characteristics within each of the major cell types in aneurysmal tissues. In this brief review, we examine the current literature utilizing scRNA-seq for the analysis of AAA and discuss trends and future utility of this technology.

5.
Int J Mol Sci ; 24(9)2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37175802

ABSTRACT

Platelet-derived growth factor type BB (PDGF-BB) regulates vascular smooth muscle cell (VSMC) migration and proliferation, which play critical roles in the development of vascular conditions. p90 ribosomal S6 kinase (p90RSK) can regulate various cellular processes through many different target substrates in several cell types, but the regulatory function of p90RSK on PDGF-BB-mediated cell migration and proliferation and subsequent vascular neointima formation has not yet been extensively examined. In this study, we investigated whether p90RSK inhibition protects VSMCs against PDGF-BB-induced cellular phenotypic changes and the molecular mechanisms underlying the effect of p90RSK inhibition on neointimal hyperplasia in vivo. Pretreatment of cultured primary rat VSMCs with FMK or BI-D1870, which are specific inhibitors of p90RSK, suppressed PDGF-BB-induced phenotypic changes, including migration, proliferation, and extracellular matrix accumulation, in VSMCs. Additionally, FMK and BI-D1870 repressed the PDGF-BB-induced upregulation of cyclin D1 and cyclin-dependent kinase-4 expression. Furthermore, p90RSK inhibition hindered the inhibitory effect of PDGF-BB on Cdk inhibitor p27 expression, indicating that p90RSK may induce VSMC proliferation by regulating the G0/G1 phase. Notably, treatment with FMK resulted in attenuation of neointima development in ligated carotid arteries in mice. The findings imply that p90RSK inhibition mitigates the phenotypic switch and neointimal hyperplasia induced by PDGF-BB.


Subject(s)
Muscle, Smooth, Vascular , Neointima , Rats , Mice , Animals , Becaplermin/pharmacology , Becaplermin/metabolism , Neointima/metabolism , Hyperplasia/metabolism , Muscle, Smooth, Vascular/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Cell Proliferation , Rats, Sprague-Dawley , Cell Movement , Myocytes, Smooth Muscle/metabolism , Cells, Cultured , Proto-Oncogene Proteins c-sis/pharmacology , Proto-Oncogene Proteins c-sis/metabolism
6.
Microvasc Res ; 149: 104554, 2023 09.
Article in English | MEDLINE | ID: mdl-37236346

ABSTRACT

Intracranial aneurysm (IA) is a severe cerebrovascular disease characterized by abnormal bulging of cerebral vessels that may rupture and cause a stroke. The expansion of the aneurysm accompanies by the remodeling of vascular matrix. It is well-known that vascular remodeling is a process of synthesis and degradation of extracellular matrix (ECM), which is highly dependent on the phenotype of vascular smooth muscle cells (VSMCs). The phenotypic switching of VSMC is considered to be bidirectional, including the physiological contractile phenotype and alternative synthetic phenotype in response to injury. There is increasing evidence indicating that VSMCs have the ability to switch to various phenotypes, including pro-inflammatory, macrophagic, osteogenic, foamy and mesenchymal phenotypes. Although the mechanisms of VSMC phenotype switching are still being explored, it is becoming clear that phenotype switching of VSMCs plays an essential role in IA formation, progression, and rupture. This review summarized the various phenotypes and functions of VSMCs associated with IA pathology. The possible influencing factors and potential molecular mechanisms of the VSMC phenotype switching were further discussed. Understanding how phenotype switching of VSMC contributed to the pathogenesis of unruptured IAs can bring new preventative and therapeutic strategies for IA.


Subject(s)
Intracranial Aneurysm , Muscle, Smooth, Vascular , Humans , Muscle, Smooth, Vascular/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/pathology , Signal Transduction , Myocytes, Smooth Muscle/pathology , Phenotype , Cells, Cultured , Cell Proliferation
7.
Mol Brain ; 15(1): 97, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36451193

ABSTRACT

Intercellular communication between vascular and nerve cells mediated by diffusible proteins has recently emerged as a critical intrinsic program for neural development. However, whether the vascular smooth muscle cell (VSMC) secretome regulates the connectivity of neural circuits remains unknown. Here, we show that conditioned medium from brain VSMC cultures enhances multiple neuronal functions, such as neuritogenesis, neuronal maturation, and survival, thereby improving circuit connectivity. However, protein denaturation by heating compromised these effects. Combined omics analyses of donor VSMC secretomes and recipient neuron transcriptomes revealed that overlapping pathways of extracellular matrix receptor signaling and adhesion molecule integrin binding mediate VSMC-dependent neuronal development. Furthermore, we found that human arterial VSMCs promote neuronal development in multiple ways, including expanding the time window for nascent neurite initiation, increasing neuronal density, and promoting synchronized firing, whereas human umbilical vein VSMCs lack this capability. These in vitro data indicate that brain arteriolar VSMCs may carry direct instructive information for neural development through intercellular communication in vivo.


Subject(s)
Brain , Neurogenesis , Humans , Biological Transport , Neurons , Myocytes, Smooth Muscle
8.
Front Cardiovasc Med ; 9: 972836, 2022.
Article in English | MEDLINE | ID: mdl-36312244

ABSTRACT

Vascular calcification (VC) is an important hallmark of cardiovascular disease, the osteo-/chondrocyte phenotype differentiation of vascular smooth muscle cells (VSMCs) is the main cause of vascular calcification. Accumulating evidence shows that mitochondrial dysfunction may ultimately be more detrimental in the VSMCs calcification. Mitochondrial participate in essential cellular functions, including energy production, metabolism, redox homeostasis regulation, intracellular calcium homeostasis, apoptosis, and signal transduction. Mitochondrial dysfunction under pathological conditions results in mitochondrial reactive oxygen species (ROS) generation and metabolic disorders, which further lead to abnormal phenotypic differentiation of VSMCs. In this review, we summarize existing studies targeting mitochondria as a treatment for VC, and focus on VSMCs, highlighting recent progress in determining the roles of mitochondrial processes in regulating the phenotype transition of VSMCs, including mitochondrial biogenesis, mitochondrial dynamics, mitophagy, mitochondrial energy metabolism, and mitochondria/ER interactions. Along these lines, the impact of mitochondrial homeostasis on VC is discussed.

9.
Biochem Biophys Res Commun ; 625: 31-37, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35944361

ABSTRACT

Dilated cardiomyopathy, a type of heart muscle disease defined by the presence of left ventricular dilatation and contractile dysfunction, is an important cause of sudden cardiac death and heart failure. O-GlcNAcylation is an important post-translational modification of proteins by the addition of O-GlcNAc moieties at serine or threonine residues. Several studies have shown that proper control of O-GlcNAcylation is required for maintaining physiological function of heart by using Ogt (O-GlcNAc transferase) cardiomyocyte-specific knockout mouse models. In this study, we generated a new mouse model (αSMA-Ogt KO) in which Ogt was deleted in both cardiomyocytes and smooth muscle cells by crossing Ogt floxed mice with αSMA-Cre mice. αSMA-Cre-mediated Ogt deletion in mice led to severe postnatal lethality; the survived mice were smaller than control mice, had dilated hearts, and showed observable signs of heart failure. Moreover, the αSMA-Ogt KO heart had more apoptotic cells and fibrosis. The arteries of αSMA-Ogt KO mice exhibited significantly reduced expression of contractile genes and a trend towards arterial stiffness. In conclusion, our data emphasize the importance of OGT in maintaining normal heart function and reveal a novel role of OGT in regulating arterial contractility.


Subject(s)
Heart Failure , Muscle, Smooth, Vascular , Animals , Heart Failure/genetics , Heart Failure/metabolism , Integrases , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , N-Acetylglucosaminyltransferases/metabolism
10.
Front Pharmacol ; 13: 836710, 2022.
Article in English | MEDLINE | ID: mdl-35153800

ABSTRACT

Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aortic wall and normally exist in a quiescent, contractile phenotype where actomyosin-derived contractile forces maintain vascular tone. However, VSMCs are not terminally differentiated and can dedifferentiate into a proliferative, synthetic phenotype. Actomyosin force generation is essential for the function of both phenotypes. Whilst much is already known about the mechanisms of VSMC actomyosin force generation, existing assays are either low throughput and time consuming, or qualitative and inconsistent. In this study, we use polyacrylamide hydrogels, tuned to mimic the physiological stiffness of the aortic wall, in a VSMC contractility assay. Isolated VSMC area decreases following stimulation with the contractile agonists angiotensin II or carbachol. Importantly, the angiotensin II induced reduction in cell area correlated with increased traction stress generation. Inhibition of actomyosin activity using blebbistatin or Y-27632 prevented angiotensin II mediated changes in VSMC morphology, suggesting that changes in VSMC morphology and actomyosin activity are core components of the contractile response. Furthermore, we show that microtubule stability is an essential regulator of isolated VSMC contractility. Treatment with either colchicine or paclitaxel uncoupled the morphological and/or traction stress responses of angiotensin II stimulated VSMCs. Our findings support the tensegrity model of cellular mechanics and we demonstrate that microtubules act to balance actomyosin-derived traction stress generation and regulate the morphological responses of VSMCs.

11.
Cells ; 10(12)2021 12 15.
Article in English | MEDLINE | ID: mdl-34944046

ABSTRACT

Activation of the type I angiotensin receptor (AT1-R) in vascular smooth muscle cells (VSMCs) plays a crucial role in the regulation of blood pressure; however, it is also responsible for the development of pathological conditions such as vascular remodeling, hypertension and atherosclerosis. Stimulation of the VSMC by angiotensin II (AngII) promotes a broad variety of biological effects, including gene expression changes. In this paper, we have taken an integrated approach in which an analysis of AngII-induced gene expression changes has been combined with the use of small-molecule inhibitors and lentiviral-based gene silencing, to characterize the mechanism of signal transduction in response to AngII stimulation in primary rat VSMCs. We carried out Affymetrix GeneChip experiments to analyze the effects of AngII stimulation on gene expression; several genes, including DUSP5, DUSP6, and DUSP10, were identified as upregulated genes in response to stimulation. Since various dual-specificity MAPK phosphatase (DUSP) enzymes are important in the regulation of mitogen-activated protein kinase (MAPK) signaling pathways, these genes have been selected for further analysis. We investigated the kinetics of gene-expression changes and the possible signal transduction processes that lead to altered expression changes after AngII stimulation. Our data shows that the upregulated genes can be stimulated through multiple and synergistic signal transduction pathways. We have also found in our gene-silencing experiments that epidermal growth factor receptor (EGFR) transactivation is not critical in the AngII-induced expression changes of the investigated genes. Our data can help us understand the details of AngII-induced long-term effects and the pathophysiology of AT1-R. Moreover, it can help to develop potential interventions for those symptoms that are induced by the over-functioning of this receptor, such as vascular remodeling, cardiac hypertrophy or atherosclerosis.


Subject(s)
Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase Phosphatases/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/enzymology , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/pharmacology , Animals , Cell Line , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Kinetics , Lentivirus/metabolism , Male , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Up-Regulation/genetics
12.
J Biol Chem ; 297(5): 101258, 2021 11.
Article in English | MEDLINE | ID: mdl-34599966

ABSTRACT

The underlying mechanism of neointima formation remains unclear. Ubiquitin-specific peptidase 10 (USP10) is a deubiquitinase that plays a major role in cancer development and progression. However, the function of USP10 in arterial restenosis is unknown. Herein, USP10 expression was detected in mouse arteries and increased after carotid ligation. The inhibition of USP10 exhibited thinner neointima in the model of mouse carotid ligation. In vitro data showed that USP10 deficiency reduced proliferation and migration of rat thoracic aorta smooth muscle cells (A7r5) and human aortic smooth muscle cells (HASMCs). Mechanically, USP10 can bind to Skp2 and stabilize its protein level by removing polyubiquitin on Skp2 in the cytoplasm. The overexpression of Skp2 abrogated cell cycle arrest induced by USP10 inhibition. Overall, the current study demonstrated that USP10 is involved in vascular remodeling by directly promoting VSMC proliferation and migration via stabilization of Skp2 protein expression.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Line , Cell Movement , Cell Proliferation , Humans , Neointima/genetics , Protein Stability , S-Phase Kinase-Associated Proteins/genetics , Ubiquitin Thiolesterase/genetics
13.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255685

ABSTRACT

BACKGROUND: Autophagy is a highly conserved catabolic homeostatic process, crucial for cell survival. It has been shown that autophagy can modulate different cardiovascular pathologies, including vascular calcification (VCN). OBJECTIVE: To assess how modulation of autophagy, either through induction or inhibition, affects vascular and valvular calcification and to determine the therapeutic applicability of inducing autophagy. DATA SOURCES: A systematic review of English language articles using MEDLINE/PubMed, Web of Science (WoS) and the Cochrane library. The search terms included autophagy, autolysosome, mitophagy, endoplasmic reticulum (ER)-phagy, lysosomal, calcification and calcinosis. Study characteristics: Thirty-seven articles were selected based on pre-defined eligibility criteria. Thirty-three studies (89%) studied vascular smooth muscle cell (VSMC) calcification of which 27 (82%) studies investigated autophagy and six (18%) studies lysosomal function in VCN. Four studies (11%) studied aortic valve calcification (AVCN). Thirty-four studies were published in the time period 2015-2020 (92%). CONCLUSION: There is compelling evidence that both autophagy and lysosomal function are critical regulators of VCN, which opens new perspectives for treatment strategies. However, there are still challenges to overcome, such as the development of more selective pharmacological agents and standardization of methods to measure autophagic flux.


Subject(s)
Aortic Valve Stenosis/genetics , Aortic Valve/pathology , Autophagy/genetics , Calcinosis/genetics , Lysosomes/genetics , Vascular Calcification/genetics , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Cell Survival/genetics , Endoplasmic Reticulum/genetics , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Vascular Calcification/metabolism , Vascular Calcification/pathology
14.
Cardiovasc Diagn Ther ; 10(4): 687-694, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32968625

ABSTRACT

BACKGROUND: The occurrence and development of atherosclerosis (AS) are closely related to the abnormality of vascular smooth muscle cells (VSMCs), and multiple microRNAs (miRNAs) have been reported to participate in the pathogenesis of AS. This study explored the expression and clinical value of miR-374 in the serum of AS patients, and analyzed its effect on the proliferation and migration of VSMCs. METHODS: The expression levels of miR-374 in the serum of 102 asymptomatic patients with AS and 89 healthy patients were detected by fluorescence quantitative PCR. The diagnostic value of miR-374 was evaluated through the receiver operating characteristic (ROC) curve. What's more, CCK-8 and Transwell assays were used to analyze the effects of miR-374 on the proliferation and migration of VSMCs. RESULTS: The expression level of miR-374 in the serum of AS patients was significantly higher than that of the control group. At the same time, the expression of miR-374 in AS patients was positively correlated with carotid intima-media thickness (CIMT). The area under the ROC curve is 0.824. Furthermore, overexpression of miR-374 significantly promoted the proliferation and migration of VSMCs, whereas reducing miR-374 inhibited the proliferation and migration of VSMCs. CONCLUSIONS: The high expression of miR-374 may be a potential diagnostic marker for AS, and overexpression of miR-374 may play a role in AS by promoting the proliferation and migration of VSMCs.

15.
Front Pharmacol ; 11: 626, 2020.
Article in English | MEDLINE | ID: mdl-32499695

ABSTRACT

Nϵ-carboxymethyl-lysine (CML), an advanced glycation end product, is involved in vascular calcification (VC) in diabetic atherosclerosis. This study aimed to investigate the effects of CML on VC in diabetic atherosclerosis induced by vascular smooth muscle cell (VSMC)-derived foam cells. Human studies, animal studies and cell studies were performed. The human study results from 100 patients revealed a poor blood glucose and lipid status and more severe coronary lesions and stenosis in patients with coronary artery disease and diabetes mellitus. Intraperitoneal injection of streptozotocin combined with a high-fat diet was used to build a diabetic atherosclerosis model in ApoE-/- mice. The animal study results indicated that CML accelerated VC progression in diabetic atherosclerosis by accelerating the accumulation of VSMC-derived foam cells in ApoE-/- mice. The cell study results illustrated that CML induced VSMC-derived foam cells apoptosis and aggravated foam cells calcification. Consistent with this finding, calcium content and the expression levels of alkaline phosphatase, bone morphogenetic protein 2 and runt-related transcription factor 2 were significantly elevated in A7r5 cells treated with oxidation-low-density lipoprotein and CML. Thus, we concluded that CML promoted VSMC-derived foam cells calcification to aggravate VC in diabetic atherosclerosis, providing evidence for the contribution of foam cells to diabetic VC.

16.
Development ; 147(10)2020 05 18.
Article in English | MEDLINE | ID: mdl-32423977

ABSTRACT

The thin endothelial wall of a newly formed vessel is under enormous stress at the onset of blood flow, rapidly acquiring support from mural cells (pericytes and vascular smooth muscle cells; vSMCs) during development. Mural cells then develop vasoactivity (contraction and relaxation) but we have little information as to when this first develops or the extent to which pericytes and vSMCs contribute. For the first time, we determine the dynamic developmental acquisition of vasoactivity in vivo in the cerebral vasculature of zebrafish. We show that pericyte-covered vessels constrict in response to α1-adrenergic receptor agonists and dilate in response to nitric oxide donors at 4 days postfertilization (dpf) but have heterogeneous responses later, at 6 dpf. In contrast, vSMC-covered vessels constrict at 6 dpf, and dilate at both stages. Using genetic ablation, we demonstrate that vascular constriction and dilation is an active response. Our data suggest that both pericyte- and vSMC-covered vessels regulate their diameter in early development, and that their relative contributions change over developmental time.


Subject(s)
Muscle, Smooth, Vascular/embryology , Myocytes, Smooth Muscle/physiology , Pericytes/physiology , Zebrafish/embryology , Zebrafish/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Animals, Genetically Modified , Brain/blood supply , Brain/diagnostic imaging , Brain/embryology , Endothelial Cells/physiology , Endothelium, Vascular/embryology , Gene Silencing , Metronidazole/pharmacology , Muscle Contraction/drug effects , Nitric Oxide Donors/pharmacology , Vasodilation/drug effects
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-817631

ABSTRACT

@#【Objective】To observe whether berberine can inhibit vascular smooth muscle cells(VSMC)proliferation induced by mechanical strength stress and to investigate the role of MAPK pathway in it.【Methods】The cultured VSMC were divided into 4 groups:negative control group(NC group),stretch stress group(SS group),berberine pretreated and stretch stress stimulation group(BBR+SS group),and berberine group. In NC group,phosphate buffer saline was used as a negative control;in SS group,stretch stress was given to VSMC;in BBR+SS group,VSMC were pretreated with berberine for 1 hour and then exposed to stretch stress;in BBR group,VSMC were treated only with berberine for 1 hour and cultured in serum- free DMEM afterwards. We collected VSMC in each group ,detected and analyzed their MAPK phosphorylation,proliferation and migration by using Western blotting,immunofluorescence and wound-healing assay respectively. 【Results】 Compare with NC group,stretch stress markedly induced VSMC proliferation and migration ,which could be inhibited significantly by berberine. Stretch stress obviously increased phosphorylation of MAPK (ERK,JNK,p38),which could be inhibited by berberine in a concentration dependent manner. 【Conclusion】 Berberine inhibits hypertension-induced proliferation and migration of VSMC through MAPK pathway. The results revealed the new use and mechanism of berberine,and provided important data for further study on the prevention and treatment of vascular remodeling caused by abnormal increase of mechanical stress in hypertension.

18.
Front Cell Dev Biol ; 7: 228, 2019.
Article in English | MEDLINE | ID: mdl-31681761

ABSTRACT

The early mammalian embryo is characterized by the presence of three germ layers-the outer ectoderm, middle mesoderm and inner endoderm. The mesoderm is organized into paraxial, intermediate and lateral plate mesoderm. The musculature, vasculature and heart of the adult body are the major derivatives of mesoderm. Tracing back the developmental process to generate these specialized tissues has sparked much interest in the field of regenerative medicine focusing on generating specialized tissues to treat patients with degenerative diseases. Several Long Non-Coding RNAs (lncRNAs) have been identified as regulators of development, proliferation and differentiation of various tissues of mesodermal origin. A better understanding of lncRNAs that can regulate the development of these tissues will open potential avenues for their therapeutic utility and enhance our knowledge about disease progression and development. In this review, we aim to summarize the functions and mechanisms of lncRNAs regulating the early mesoderm differentiation, development and homeostasis of skeletal muscle and cardiovascular system with an emphasis on their therapeutic potential.

19.
Biol Pharm Bull ; 42(11): 1783-1788, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31391347

ABSTRACT

To examine the effect of tanshinone IIA on Angiotensin II (Ang II)-induced proliferation and autophagy in vascular smooth muscle cells (VSMCs) and the related mechanism. VSMCs were treated with Ang II with or without tanshinone IIA (1, 5 and 10 µg/mL), and the proliferation, apoptosis in cells with different treatment were examined by methylthiazolyl tetrazolium (MTT) and flow cytometry methods. Moreover, the expression of autophagy related proteins and mitogen-activated protein kinase (MAPK) signaling molecules were examined by RT-quantitative (q)PCR and Western blot methods. Ang II induced significantly increase in the proliferation and autophagy of VSMCs, and the MAPK signaling was activated. Tanshinone IIA can attenuate Ang II-induced effects via down-regulating the MAPK signaling pathway. Tanshinone IIA can inhibit Ang II-induced proliferation and autophagy of VSMCs via regulating the MAPK signaling pathway.


Subject(s)
Abietanes/pharmacology , Angiotensin II/metabolism , Autophagy/drug effects , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/metabolism , Animals , Cells, Cultured , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Primary Cell Culture , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Mol Reprod Dev ; 86(6): 686-695, 2019 06.
Article in English | MEDLINE | ID: mdl-31066488

ABSTRACT

Extravillous trophoblasts (EVTs) migrate into uterine decidua and induce vascular smooth muscle cell (VSMC) loss through mechanisms thought to involve migration and apoptosis, achieving complete spiral artery remodeling. Long noncoding RNA maternally expressed gene 3 (MEG3) can regulate diverse cellular processes, such as proliferation and migration, and has been discovered highly expressed in human placenta tissues. However, little is known about the role of MEG3 in modulating EVT functions and EVT-induced VSMC loss. In this study, we first examined the location of MEG3 in human first-trimester placenta by in situ hybridization. Then, exogenous upregulation of MEG3 in HTR-8/SVneo cells was performed to investigate the effects of MEG3 on EVT motility and EVT capacity to displace VSMCs. Meanwhile, the molecules mediating EVT-induced VSMC loss, such as tumor necrosis factor-α (TNF-α), Fas ligand (FasL), and tumor necrosis factor-α-related apoptosis-inducing ligand (TRAIL) were detected at transcriptional and translational levels. Finally, VSMCs were cocultured with MEG3-upregulated HTR-8/SVneo to explore the role of MEG3 on EVT-mediated VSMC migration and apoptosis. Results showed that MEG3 was expressed in trophoblasts in placental villi and decidua, and MEG3 enhancement inhibited HTR-8/SVneo migration and invasion. Meanwhile, the displacement of VSMCs by HTR-8/SVneo and the expression of TNF-α, FasL and TRAIL in HTR-8/SVneo were reduced following MEG3 overexpression in HTR-8/SVneo. Furthermore, HTR-8/SVneo with MEG3 upregulation impaired VSMC migration and apoptosis. The PI3K/Akt pathway, which is possibly downstream, was inactivated in MEG3-upregulated HTR-8/SVneo. These findings suggest that MEG3 might be a negative regulator of spiral artery remodeling via suppressing EVT invasion and EVT-mediated VSMC loss.


Subject(s)
Cell Movement , Decidua/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Long Noncoding/metabolism , Trophoblasts/metabolism , Uterine Artery/metabolism , Vascular Remodeling , Cell Line , Decidua/cytology , Female , Humans , Muscle, Smooth, Vascular/cytology , Pregnancy , Pregnancy Trimester, First , Trophoblasts/cytology , Uterine Artery/cytology
SELECTION OF CITATIONS
SEARCH DETAIL