Abstract 133: Mechanistic Insights Into Bradykinin and Thromboxane Receptors Heterodimerization in Vascular Smooth Muscle Cells

2016 ◽  
Vol 36 (suppl_1) ◽  
Author(s):  
Oula K Dagher ◽  
Miran A Jaffa ◽  
Aïda Habib ◽  
Fuad N Ziyadeh ◽  
Louis M Luttrell ◽  
...  

The development of an atherosclerotic lesion in an injured vasculature is highly dependent on the proliferative state of vascular smooth muscle cells (VSMC). Bradykinin (BK) and Thromboxane are two G-protein coupled receptor ligands, whose individual binding to their respective receptors, B 2 R and TP, promotes ERK1/2-mediated VSMC proliferation. However, it is not yet known whether receptor-receptor interactions between B 2 R and TP could contribute to their co-regulation. Thus, our work addresses the hypothesis that, in VSMC, B 2 R and TP form functional hetero-complexes, of distinct trafficking and signaling properties. B 2 R-TP signaling crosstalk was first analyzed in rat VSMC by Western Blot analysis and subsequent fold/basal quantification of ERK1/2 phosphorylation. B 2 R-TP cooperation was evident through the synergistic ERK1/2 phosphorylation, in VSMC co-stimulated with optimized combinations of BK and the TP agonist, IBOP (21.05 ± 4.93 fold of basal, n=3; p < 0.001). Interestingly, however, pretreatment with the TP antagonist, SQ29548, totally abolished (BK+IBOP)-induced ERK1/2 (n=3; p < 0.001). Furthermore, knowing that B 2 R, unlike the human TPα isoform, undergoes agonist-induced sequestration and β-arrestin2 recruitment, we conducted immunofluorescence analysis on HEK293T cells overexpressing human B 2 R and human TPα (B 2 R-TPα-HEKs). While stimulation with IBOP failed to mobilize cell membranous B 2 R or TPα, substantial co-internalization of TPα and B 2 R was seen, subsequent to stimulation of B 2 R-TPα-HEKs with BK. Likewise, overexpressing β-arrestin2 in B 2 R-TPα-HEKs resulted in co-localization of B 2 R, TPα, and β-arrestin2 within internalized puncta, only after BK stimulation. Pretreatment with SQ29548 inhibited BK-induced co-internalization and β-arrestin2 recruitment. However, SQ29548 could not reverse BK-induced B 2 R sequestration in HEK293T cells overexpressing B 2 R alone, thus excluding the possibility of direct SQ29548 - B 2 R binding. Finally, results of our ongoing work are the first to show nuclear localization of B 2 R and TP within human and rat VSMC. Thus, our findings favor the likelihood of functional B 2 R-TP heterodimerization in VSMC, which could serve as a novel target for interventional strategies.

2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Baoliang Zhu ◽  
Jing Liu ◽  
Ying Zhao ◽  
Jing Yan

Coronary heart disease (CHD) is the most common cardiovascular disease with high prevalence, disability, and mortality. The balance between proliferation and apoptosis of vascular smooth muscle cells (VSMCs) plays a key role in the initiation of atherosclerosis. In this study, we found a significant decrease in the expression of lncRNA-SNHG14 in atherosclerotic plaque tissues of ApoE-/- mice. Overexpression of lncRNA-SNHG14 can inhibit VSMC proliferation while promoting apoptosis. There is a potential reciprocal regulatory relationship between lncRNASNHG14 and miR-19a-3p, which inhibit each other’s expression in vascular smooth muscle cells. In addition, the luciferase reporter gene analysis results showed that there was a direct interaction between miR-19a-3p and the 3′UTR of RORα. The results of qRT-PCR showed that the level of RORα mRNA was significantly increased in the aortas treated with miR-19a-3p and SNHG14 compared with that treated with miR-19a-3p alone. In conclusion, we demonstrated that lncRNA-SNHG14 regulates the apoptosis/proliferation balance of VSMCs in atherosclerosis.


2020 ◽  
Vol 72 (2) ◽  
pp. 165-172 ◽  
Author(s):  
Lei Li ◽  
Yilin Xie ◽  
Shen Li ◽  
Juanjuan Tan ◽  
Yingchun Qin ◽  
...  

Cytoglobin, a recently discovered globin, is expressed in vascular smooth muscle cells (VSMCs). Loss of cytoglobin provides a protective effect on vascular reconstruction but the effect of its overexpression is unclear. The aim of the study was to investigate the effect of cytoglobin overexpression on the migration and proliferation of VSMCs and possible mechanisms. We detected the expression of cytoglobin in hypertensive and normotensive rat aortas, with negative feedback regulation between cytoglobin and hypertension observed. The expression of cytoglobin was significantly decreased in hypertensive rats compared to normotensive rats, but VSMCs overexpressing cytoglobin displayed increased cell migration and proliferation, which led to a phenotypic switch. The increased expression of matrix metalloproteinase 9 and collagen Ia suggests a role for cytoglobin in extracellular matrix remodeling. Increased expression of proliferating cell nuclear antigen and decreased expression of p27 implies that cytoglobin is involved in modulating VSMC proliferation. Our findings indicate that cytoglobin may play an important role in vascular wall remodeling.


Vascular ◽  
2020 ◽  
Vol 28 (6) ◽  
pp. 821-828
Author(s):  
Hong-Xia Tang ◽  
Xu-Ping Qin ◽  
Jie Li

Objectives Cardiovascular disease (CVD) remains the primary cause of morbidity and mortality worldwide. The abnormal proliferation of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of CVD. The functional and phenotypic changes in vascular cells are mediated by complex signaling cascades that initiate and control genetic reprogramming. Many studies have demonstrated that signal transducer and activator of transcription 3 (STAT3) regulates a diverse array of functions relevant to atherosclerosis. Methods In this review, we summarize the studies on the STAT3-mediated proliferation of VSMCs and subsequent CVDs such as hypertension, atherosclerosis, stroke, coronary artery disease, and myocardial infarction. Furthermore, we describe the general background of STAT3, its structure, function and regulation as well as the STAT3 signaling pathway. Finally, we highlight some potential issues and propose some solutions to these issues. Results and conclusions: STAT3 activation promotes the proliferation of VSMCs by regulating the transcription of genes. Studying the mechanism of VSMC proliferation induced by the STAT3 pathway is valuable for finding therapeutic targets for CVD.


2018 ◽  
Vol 19 (2) ◽  
pp. 153-161 ◽  
Author(s):  
Liqin Yuan ◽  
Chang Shu ◽  
Xiao Zhou ◽  
Jiehua Li ◽  
Lunchang Wang ◽  
...  

Purpose: To study the effect of x-ray radiotherapy on vascular smooth muscle cells (VSMCs) and elucidate the mechanisms in preventing neointimal hyperplasia of prosthetic vascular grafts. Materials and methods: In model I, twelve mongrel dogs underwent revascularization with prosthetic grafts and half the dogs underwent irradiation of the grafts at 28 Gy. In model II, human VSMCs (hVSMCs) were maintained and divided into six groups to which external radiation was applied at six different doses: 0 Gy, 2 Gy, 8 Gy, 16 Gy, 24 Gy and 30 Gy. In both models, specimens were harvested and examined by using morphological, immunological, cellular and molecular methods. Results: After irradiation, the neointima thickness was significantly lower in irradiated groups (p≤0.01). The radiotherapy could up-regulate p27kip1, and down-regulate proliferating cell nuclear antigen (PCNA) and S phase kinase associated protein 2 (Skp2). X-ray irradiation inhibits the proliferation of hVSMCs via acting on G1/S phase of cell cycle. The apoptosis of hVSMCs increased significantly with dose and time. The expression of PCNA and Skp2 were decreased after a first increasing trend with dose, but had a significant negative correlation with time. The expression of p27kip1 had a significant positive correlation with dose and time. Conclusions: Postoperative external fractionated irradiation after prosthetic vessel replacement of the abdominal aorta suppressed the development of hyperplasia in the graft neointima in the short term. There was a prominent time- and dose-dependent inhibition of VSMC proliferation by radiation when it was administered.


2013 ◽  
Vol 2013 ◽  
pp. 1-10 ◽  
Author(s):  
Sun Mi Hwang ◽  
Yun Jung Lee ◽  
Yong Pyo Lee ◽  
Jung Joo Yoon ◽  
So Min Lee ◽  
...  

The abnormal proliferation of vascular smooth muscle cells (VSMCs) in arterial walls is an important pathogenic factor of vascular disorders such as diabetic atherosclerosis. We have reported the anti-inflammatory effect of an aqueous extract fromPrunella vulgaris(APV) in vascular endothelial cell. In the present study, APV exhibited inhibitory effects on high glucose-stimulated VSMC proliferation, migration, and invasion activities, inducing G1cell cycle arrest with downregulation of cyclins and CDKs and upregulation of the CKIs,p21waf1/cip1andp27kip1. Furthermore, APV dose dependently suppressed the high glucose-induced matrix metalloproteinase activity. High glucose-induced phosphorylation of ERK, p38 MAPK, was decreased by the pretreatment of APV. NF-κB activation by high glucose was attenuated by APV, as an antioxidant. APV attenuated the high glucose-induced decrease of nuclear factor E2-related factor-2 (Nrf2) translocation and heme oxygenase-1 (HO-1) expression. Intracellular cGMP level was also increased by APV treatment. These results demonstrate that APV may inhibit VSMC proliferation via downregulating ROS/NF-κB /ERK/p38 MAPK pathways. In addition, APV has a beneficial effect by the interaction of Nrf2-mediated NO/cGMP with HO-1, suggesting thatPrunella vulgarismay be useful in preventing diabetic atherosclerosis.


2011 ◽  
Vol 286 (41) ◽  
pp. 35485-35493 ◽  
Author(s):  
Florence Gizard ◽  
Yue Zhao ◽  
Hannes M. Findeisen ◽  
Hua Qing ◽  
Dianne Cohn ◽  
...  

Members of the NR4A subgroup of the nuclear hormone receptor superfamily have emerged as key transcriptional regulators of proliferation and inflammation. NOR1 constitutes a ligand-independent transcription factor of this subgroup and induces cell proliferation; however, the transcriptional mechanisms underlying this mitogenic role remain to be defined. Here, we demonstrate that the F-box protein SKP2 (S phase kinase-associated protein 2), the substrate-specific receptor of the ubiquitin ligase responsible for the degradation of p27KIP1 through the proteasome pathway, constitutes a direct transcriptional target for NOR1. Mitogen-induced Skp2 expression is silenced in vascular smooth muscle cells (VSMC) isolated from Nor1-deficient mice or transfected with Nor1 siRNA. Conversely, adenovirus-mediated overexpression of NOR1 induces Skp2 expression in VSMC and decreases protein abundance of its target p27. Transient transfection experiments establish that NOR1 transactivates the Skp2 promoter through a nerve growth factor-induced clone B response element (NBRE). Electrophoretic mobility shift and chromatin immunoprecipitation assays further revealed that NOR1 is recruited to this NBRE site in the Skp2 promoter in response to mitogenic stimulation. In vivo Skp2 expression is increased during the proliferative response underlying neointima formation, and this transcriptional induction depends on the expression of NOR1. Finally, we demonstrate that overexpression of Skp2 rescues the proliferative arrest of Nor1-deficient VSMC. Collectively, these results characterize Skp2 as a novel NOR1-regulated target gene and detail a previously unrecognized transcriptional cascade regulating mitogen-induced VSMC proliferation.


1992 ◽  
Vol 283 (2) ◽  
pp. 403-408 ◽  
Author(s):  
D J Grainger ◽  
T R Hesketh ◽  
P L Weissberg ◽  
J C Metcalfe

Hexamethylenebisacetamide (HMBA) selectively and reversibly inhibited proliferation of human and rat vascular smooth-muscle cells (VSMCs) compared with endothelial cells, fibroblasts or lymphocytes. Half-maximal inhibition of VSMC proliferation occurred at 2-5 mM-HMBA, and at 30- greater than 50 mM for other cell types. HMBA also prevented de-differentiation, defined by the loss of smooth-muscle-specific myosin heavy chain, of primary rat VSMCs and caused partial re-differentiation of subcultured cells. Other inhibitors of ADP-ribosyltransferase were also selective inhibitors of VSMC proliferation.


2004 ◽  
Vol 287 (6) ◽  
pp. H2376-H2383 ◽  
Author(s):  
Xiaohong Wang ◽  
Neeta Adhikari ◽  
Qinglu Li ◽  
Jennifer L. Hall

Initial studies have established expression of low-density lipoprotein (LDL) receptor-related protein 6 (LRP6) in vascular smooth muscle cells (VSMCs). We hypothesized that LRP6 is a critical mediator governing the regulation of the canonical Wnt/β-catenin/T cell factor 4 (Tcf-4) cascade in the vasculature. This hypothesis was based on our previous work demonstrating a role for the β-catenin/Tcf-4 pathway in vascular remodeling as well as work in other cell systems establishing a role for LRP family members in the Wnt cascade. In line with our hypothesis, LRP6 upregulation significantly increased Wnt-1-induced Tcf activation. Moreover, a dominant interfering LRP6 mutant lacking the carboxyl intracellular domain (LRP6ΔC) abolished Tcf activity. LRP6-induced stimulation of Tcf was blocked in VSMCs harboring constitutive expression of a dominant negative Tcf-4 transgene lacking the β-catenin binding domain, suggesting that LRP6-induced activation of Tcf was mediated through a β-catenin-dependent signal. Expression of the dominant interfering LRP6ΔC transgene was sufficient to abolish the Wnt-induced survival as well as cyclin D1 activity and cell cycle progression. In conclusion, these findings provide the first evidence of a role for an LDL receptor-related protein in the regulation of VSMC proliferation and survival through the evolutionary conserved Wnt signaling cascade.


2021 ◽  
Author(s):  
Mandy O J Grootaert ◽  
Martin R Bennett

Abstract Vascular smooth muscle cells (VSMCs) are key participants in both early and late-stage atherosclerosis. VSMCs invade the early atherosclerotic lesion from the media, expanding lesions, but also forming a protective fibrous cap rich in extracellular matrix to cover the ‘necrotic’ core. Hence, VSMCs have been viewed as plaque-stabilizing, and decreased VSMC plaque content—often measured by expression of contractile markers—associated with increased plaque vulnerability. However, the emergence of lineage-tracing and transcriptomic studies has demonstrated that VSMCs comprise a much larger proportion of atherosclerotic plaques than originally thought, demonstrate multiple different phenotypes in vivo, and have roles that might be detrimental. VSMCs down-regulate contractile markers during atherosclerosis whilst adopting alternative phenotypes, including macrophage-like, foam cell-like, osteochondrogenic-like, myofibroblast-like, and mesenchymal stem cell-like. VSMC phenotypic switching can be studied in tissue culture, but also now in the media, fibrous cap and deep-core region, and markedly affects plaque formation and markers of stability. In this review, we describe the different VSMC plaque phenotypes and their presumed cellular and paracrine functions, the regulatory mechanisms that control VSMC plasticity, and their impact on atherogenesis and plaque stability.


Sign in / Sign up

Export Citation Format

Share Document