Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.699
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000253

ABSTRACT

It has been reported that, in the spontaneously hypertensive rat (SHR) model of hypertension, different components of the G-protein/adenylate cyclase (AC)/Calcium-activated potassium channel of high conductance (BK) channel signaling pathway are altered differently. In the upstream part of the pathway (G-protein/AC), a comparatively low efficacy has been established, whereas downstream BK currents seem to be increased. Thus, the overall performance of this signaling pathway in SHR is elusive. For a better understanding, we focused on one aspect, the direct targeting of the BK channel by the G-protein/AC pathway and tested the hypothesis that the comparatively low AC pathway efficacy in SHR results in a reduced agonist-induced stimulation of BK currents. This hypothesis was investigated using freshly isolated smooth muscle cells from WKY and SHR rat tail artery and the patch-clamp technique. It was observed that: (1) single BK channels have similar current-voltage relationships, voltage-dependence and calcium sensitivity; (2) BK currents in cells with a strong buffering of the BK channel activator calcium have similar current-voltage relationships; (3) the iloprost-induced concentration-dependent increase of the BK current is larger in WKY compared to SHR; (4) the effects of activators of the PKA pathway, the catalytic subunit of PKA and the potent and selective cAMP-analogue Sp-5,6-DCl-cBIMPS on BK currents are similar. Thus, our data suggest that the lower iloprost-induced stimulation of the BK current in freshly isolated rat tail artery smooth muscle cells from SHR compared with WKY is due to the lower efficacy of upstream elements of the G-Protein/AC/BK channel pathway.


Subject(s)
Calcium , Hypertension , Iloprost , Large-Conductance Calcium-Activated Potassium Channels , Muscle, Smooth, Vascular , Rats, Inbred SHR , Rats, Inbred WKY , Vasodilator Agents , Animals , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Rats , Calcium/metabolism , Iloprost/pharmacology , Hypertension/metabolism , Hypertension/drug therapy , Vasodilator Agents/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Male , Arteries/drug effects , Arteries/metabolism , Tail/blood supply , Signal Transduction/drug effects
2.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000533

ABSTRACT

Vascular calcification (VC) is a cardiovascular disease characterized by calcium salt deposition in vascular smooth muscle cells (VSMCs). Standard in vitro models used in VC investigations are based on VSMC monocultures under static conditions. Although these platforms are easy to use, the absence of interactions between different cell types and dynamic conditions makes these models insufficient to study key aspects of vascular pathophysiology. The present study aimed to develop a dynamic endothelial cell-VSMC co-culture that better mimics the in vivo vascular microenvironment. A double-flow bioreactor supported cellular interactions and reproduced the blood flow dynamic. VSMC calcification was stimulated with a DMEM high glucose calcification medium supplemented with 1.9 mM NaH2PO4/Na2HPO4 (1:1) for 7 days. Calcification, cell viability, inflammatory mediators, and molecular markers (SIRT-1, TGFß1) related to VSMC differentiation were evaluated. Our dynamic model was able to reproduce VSMC calcification and inflammation and evidenced differences in the modulation of effectors involved in the VSMC calcified phenotype compared with standard monocultures, highlighting the importance of the microenvironment in controlling cell behavior. Hence, our platform represents an advanced system to investigate the pathophysiologic mechanisms underlying VC, providing information not available with the standard cell monoculture.


Subject(s)
Cell Differentiation , Coculture Techniques , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Vascular Calcification , Humans , Vascular Calcification/metabolism , Vascular Calcification/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Cells, Cultured , Cell Survival , Transforming Growth Factor beta1/metabolism , Sirtuin 1/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Bioreactors
3.
PLoS One ; 19(7): e0303472, 2024.
Article in English | MEDLINE | ID: mdl-38990864

ABSTRACT

Plasmid transfection in cells is widely employed to express exogenous proteins, offering valuable mechanistic insight into their function(s). However, plasmid transfection efficiency in primary vascular endothelial cells (ECs) and smooth muscle cells (SMCs) is restricted with lipid-based transfection reagents such as Lipofectamine. The STING pathway, activated by foreign DNA in the cytosol, prevents foreign gene expression and induces DNA degradation. To address this, we explored the potential of STING inhibitors on the impact of plasmid expression in primary ECs and SMCs. Primary human aortic endothelial cells (HAECs) were transfected with a bicistronic plasmid expressing cytochrome b5 reductase 4 (CYB5R4) and enhanced green fluorescent protein (EGFP) using Lipofectamine 3000. Two STING inhibitors, MRT67307 and BX795, were added during transfection and overnight post-transfection. As a result, MRT67307 significantly enhanced CYB5R4 and EGFP expression, even 24 hours after its removal. In comparison, MRT67307 pretreatment did not affect transfection, suggesting the inhibitor's effect was readily reversible. The phosphorylation of endothelial nitric oxide synthase (eNOS) at Serine 1177 (S1177) by vascular endothelial growth factor is essential for endothelial proliferation, migration, and survival. Using the same protocol, we transfected wild-type and phosphorylation-incapable mutant (S1177A) eNOS in HAECs. Both forms of eNOS localized on the plasma membrane, but only the wild-type eNOS was phosphorylated by vascular endothelial growth factor treatment, indicating normal functionality of overexpressed proteins. MRT67307 and BX795 also improved plasmid expression in human and rat aortic SMCs. In conclusion, this study presents a modification enabling efficient plasmid transfection in primary vascular ECs and SMCs, offering a favorable approach to studying protein function(s) in these cell types, with potential implications for other primary cell types that are challenging to transfect.


Subject(s)
Endothelial Cells , Membrane Proteins , Plasmids , Transfection , Humans , Plasmids/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Animals , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Cells, Cultured , Phosphorylation , Rats , Gene Expression , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism
4.
Elife ; 132024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985140

ABSTRACT

Vascular smooth muscle cells (VSMCs) envelop vertebrate brain arteries and play a crucial role in regulating cerebral blood flow and neurovascular coupling. The dedifferentiation of VSMCs is implicated in cerebrovascular disease and neurodegeneration. Despite its importance, the process of VSMC differentiation on brain arteries during development remains inadequately characterized. Understanding this process could aid in reprogramming and regenerating dedifferentiated VSMCs in cerebrovascular diseases. In this study, we investigated VSMC differentiation on zebrafish circle of Willis (CoW), comprising major arteries that supply blood to the vertebrate brain. We observed that arterial specification of CoW endothelial cells (ECs) occurs after their migration from cranial venous plexus to form CoW arteries. Subsequently, acta2+ VSMCs differentiate from pdgfrb+ mural cell progenitors after they were recruited to CoW arteries. The progression of VSMC differentiation exhibits a spatiotemporal pattern, advancing from anterior to posterior CoW arteries. Analysis of blood flow suggests that earlier VSMC differentiation in anterior CoW arteries correlates with higher red blood cell velocity and wall shear stress. Furthermore, pulsatile flow induces differentiation of human brain PDGFRB+ mural cells into VSMCs, and blood flow is required for VSMC differentiation on zebrafish CoW arteries. Consistently, flow-responsive transcription factor klf2a is activated in ECs of CoW arteries prior to VSMC differentiation, and klf2a knockdown delays VSMC differentiation on anterior CoW arteries. In summary, our findings highlight blood flow activation of endothelial klf2a as a mechanism regulating initial VSMC differentiation on vertebrate brain arteries.


Subject(s)
Cell Differentiation , Circle of Willis , Hemodynamics , Muscle, Smooth, Vascular , Zebrafish , Animals , Circle of Willis/embryology , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Humans , Myocytes, Smooth Muscle/physiology , Myocytes, Smooth Muscle/metabolism , Endothelial Cells/physiology , Endothelial Cells/metabolism
5.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000009

ABSTRACT

Yohimbine (YHB) has been reported to possess anti-inflammatory, anticancer, and cardiac function-enhancing properties. Additionally, it has been reported to inhibit the proliferation, migration, and neointimal formation of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor (PDGF) stimulation by suppressing the phospholipase C-gamma 1 pathway. However, the transcriptional regulatory mechanism of YHB controlling the behavior of VSMCs is not fully understood. In this study, YHB downregulated the expression of cell cycle regulatory proteins, such as proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin-dependent kinase 4 (CDK4), and cyclin E, by modulating the transcription factor FOXO3a in VSMCs induced by PDGF. Furthermore, YHB decreased p-38 and mTOR phosphorylation in a dose-dependent manner. Notably, YHB significantly reduced the phosphorylation at Y397 and Y925 sites of focal adhesion kinase (FAK), and this effect was greater at the Y925 site than Y397. In addition, the expression of paxillin, a FAK-associated protein known to bind to the Y925 site of FAK, was significantly reduced by YHB treatment in a dose-dependent manner. A pronounced reduction in the migration and proliferation of VSMCs was observed following co-treatment of YHB with mTOR or p38 inhibitors. In conclusion, this study shows that YHB inhibits the PDGF-induced proliferation and migration of VSMCs by regulating the transcription factor FOXO3a and the mTOR/p38/FAK signaling pathway. Therefore, YHB may be a potential therapeutic candidate for preventing and treating cardiovascular diseases such as atherosclerosis and vascular restenosis.


Subject(s)
Cell Movement , Cell Proliferation , Forkhead Box Protein O3 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Platelet-Derived Growth Factor , Yohimbine , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Forkhead Box Protein O3/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Animals , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Yohimbine/pharmacology , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Cells, Cultured , Paxillin/metabolism , Rats, Sprague-Dawley , Male
6.
Tissue Cell ; 89: 102440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002288

ABSTRACT

Abnormal proliferation, migration, and foam cell formation of Vascular smooth muscle cells (VSMCs) each play a role in the development of atherosclerosis (AS). Schisandrin (Sch) is the active lignan ingredient with broad-spectrum pharmacological effects. However, the role of Sch in the AS process is not clear. Therefore, this study was proposed to explore the therapeutic effect and potential mechanism of Sch on VSMCs. Ox-LDL was selected to create an atherosclerosis injury environment for VSMCs and macrophages. The MTT assay, Oil red O staining, wound healing, transwell experiments and ELISA were used to investigate the phenotype effects of Sch. Network pharmacology, molecular docking, flow cytometry, and western blot were used to investigate the underlying mechanisms of Sch on AS progression. Our findings implied that Sch treatment inhibited the proliferation and migration of VSMCs, and suppressed the ROS production and inflammatory cytokines up-regulation of VSMCs and macrophages. Moreover, Sch reduced lipid uptake and foam cell formation through downregulating LOX-1. Mechanistically, we found that Sch can inhibit the activation of JAK2/STAT3 signaling by targeting JAK2, and arrest cell cycle in GO/G1 phase. In summary, Sch can inhibit VSMCs proliferation and migration by arresting cell cycle and targeting JAK2 to regulating the JAK2/STAT3 pathway. Sch may serve as a potential drug for patients with AS.


Subject(s)
Cell Movement , Cell Proliferation , Cyclooctanes , Janus Kinase 2 , Lignans , Muscle, Smooth, Vascular , Polycyclic Compounds , STAT3 Transcription Factor , Signal Transduction , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/cytology , Lignans/pharmacology , Signal Transduction/drug effects , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Cell Cycle Checkpoints/drug effects , Animals , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/drug therapy
7.
Int Heart J ; 65(4): 738-747, 2024.
Article in English | MEDLINE | ID: mdl-39085113

ABSTRACT

The regulatory factor X7 (RFX7) is a vital mediator in atherosclerosis. This study aims to discuss the effect and underlying mechanism of RFX7 on the regulation of oxidized low-density lipoprotein (ox-LDL) -induced proliferation and migration of vascular smooth muscle cells (VSMCs).Ox-LDL was used to construct atherosclerosis in vitro model. The mRNA and protein levels of RFX7 and Sirtuin 4 (SIRT4) were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assays. The cellular functions were measured via 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), EdU, flow cytometry, and wound healing assay assays. The interaction between RFX7 and SIRT4 promoter was validated using chromatin immunoprecipitation and dual-luciferase reporter assays.The stimulation with ox-LDL elevated the viability of VSMCs and decreased the mRNA and protein levels of RFX7 and SIRT4 in VSMCs in a dose-dependent manner. Functionally, RFX7 overexpression restrained the VSMC viability, proliferation, and migration induced by ox-LDL, but facilitated VSMC apoptosis. RFX7 elevated SIRT4 expression via binding to its promoter. Furthermore, overexpressing either SIRT4 or RFX7 inactivated JAK2/STAT3 signaling, causing a decrease in VSMC proliferation and migration and an increase in VSMC apoptosis when exposed to ox-LDL. The impact of RFX7 overexpression on JAK2/STAT3 signaling and cellular function following ox-LDL exposure was abrogated by SIRT4 silencing.The heightened RFX7 expression restrained the proliferation and migration of ox-LDL-stimulated VSMCs via SIRT4-mediated inactivation of JAK2/STAT3 pathway.


Subject(s)
Cell Movement , Cell Proliferation , Janus Kinase 2 , Lipoproteins, LDL , Muscle, Smooth, Vascular , STAT3 Transcription Factor , Signal Transduction , Sirtuins , STAT3 Transcription Factor/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , Janus Kinase 2/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Humans , Sirtuins/metabolism , Sirtuins/genetics , Atherosclerosis/metabolism , Cells, Cultured , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Mitochondrial Proteins
8.
Sci Rep ; 14(1): 16323, 2024 07 15.
Article in English | MEDLINE | ID: mdl-39009669

ABSTRACT

Vascular calcification, which is a major complication of diabetes mellitus, is an independent risk factor for cardiovascular disease. Osteogenic differentiation of vascular smooth muscle cells (VSMCs) is one of the key mechanisms underlying vascular calcification. Emerging evidence suggests that macrophage-derived extracellular vesicles (EVs) may be involved in calcification within atherosclerotic plaques in patients with diabetes mellitus. However, the role of macrophage-derived EVs in the progression of vascular calcification is largely unknown. In this study, we investigated whether macrophage-derived EVs contribute to the osteogenic differentiation of VSMCs under high glucose conditions. We isolated EVs that were secreted by murine peritoneal macrophages under normal glucose (EVs-NG) or high glucose (EVs-HG) conditions. miRNA array analysis in EVs from murine macrophages showed that miR-17-5p was significantly increased in EVs-HG compared with EVs-NG. Prediction analysis with miRbase identified transforming growth factor ß receptor type II (TGF-ß RII) as a potential target of miR-17-5p. EVs-HG as well as miR-17-5p overexpression with lipid nanoparticles inhibited the gene expression of Runx2, and TGF-ß RII. Furthermore, we demonstrated that VSMCs transfected with miR-17-5p mimic inhibited calcium deposition. Our findings reveal a novel role of macrophage-derived EVs in the negative regulation of osteogenic differentiation in VSMCs under high glucose conditions.


Subject(s)
Cell Differentiation , Extracellular Vesicles , Glucose , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Osteogenesis , Signal Transduction , Transforming Growth Factor beta , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Glucose/pharmacology , Glucose/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Transforming Growth Factor beta/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Extracellular Vesicles/metabolism , Vascular Calcification/metabolism , Vascular Calcification/genetics , Vascular Calcification/pathology , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Male , Mice, Inbred C57BL , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics
9.
J Mech Behav Biomed Mater ; 157: 106639, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38970943

ABSTRACT

An intricate reciprocal relationship exists between adherent synthetic cells and their extracellular matrix (ECM). These cells deposit, organize, and degrade the ECM, which in turn influences cell phenotype via responses that include sensitivity to changes in the mechanical state that arises from changes in external loading. Collagen-based tissue equivalents are commonly used as simple but revealing model systems to study cell-matrix interactions. Nevertheless, few quantitative studies report changes in the forces that the cells establish and maintain in such gels under dynamic loading. Moreover, most prior studies have been limited to uniaxial experiments despite many soft tissues, including arteries, experiencing multiaxial loading in vivo. To begin to close this gap, we use a custom biaxial bioreactor to subject collagen gels seeded with primary aortic smooth muscle cells to different biaxial loading conditions. These conditions include cyclic loading with different amplitudes as well as different mechanical constraints at the boundaries of a cruciform sample. Irrespective of loading amplitude and boundary condition, similar mean steady-state biaxial forces emerged across all tests. Additionally, stiffness-force relationships assessed via intermittent equibiaxial force-extension tests showed remarkable similarity for ranges of forces to which the cells adapted during periods of cyclic loading. Taken together, these findings are consistent with a load-mediated homeostatic response by vascular smooth muscle cells.


Subject(s)
Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Animals , Biomechanical Phenomena , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Tissue Engineering , Stress, Mechanical , Mechanical Phenomena , Collagen/metabolism , Bioreactors , Weight-Bearing , Extracellular Matrix/metabolism , Rats , Materials Testing , Aorta/cytology
10.
Cell Death Dis ; 15(6): 402, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851795

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation, migration, and apoptosis play important roles in many physiological processes and pathological conditions. To identify genetic influences on VSMC behavior, we measured these traits and undertook genome-wide association studies in primary umbilical artery-derived VSMCs from >2000 individuals. Although there were no genome-wide significant associations for VSMC proliferation or migration, genetic variants at two genomic loci (7p15.3 and 7q32.3) showed highly significant associations with VSMC apoptosis (P = 1.95 × 10-13 and P = 7.47 × 10-9, respectively). The lead variant at the 7p51.3 locus was associated with increased expression of the GSDME and PALS2 genes in VSMCs. Knockdown of GSDME or PALS2 in VSMCs attenuated apoptotic cell death. A protein co-immunoprecipitation assay indicated that GSDME complexed with PALS2. PALS2 knockdown attenuated activated caspase-3 and GSDME fragmentation, whilst GSDME knockdown also reduced activated caspase-3. These findings provide new insights into the genetic regulation of VSMC apoptosis, with potential utility for therapeutic development.


Subject(s)
Apoptosis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Apoptosis/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Humans , Myocytes, Smooth Muscle/metabolism , Genome-Wide Association Study , Caspase 3/metabolism , Caspase 3/genetics , Cell Proliferation/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Movement/genetics , Cells, Cultured
12.
Int Heart J ; 65(3): 557-565, 2024.
Article in English | MEDLINE | ID: mdl-38825498

ABSTRACT

When stimulated, vascular smooth muscle cells (VSMCs) change from a differentiated to a dedifferentiated phenotype. Dedifferentiated VSMCs have a key activity in cardiovascular diseases such as in-stent restenosis. MicroRNAs (miRNAs) have crucial functions in conversion of differentiated VSMCs to a dedifferentiated phenotype. We investigated the activity of miR-411-5p in the proliferation, migration, and phenotype switch of rat VSMCs.Based on a microRNA array assay, miR-411-5p expression was found to be significantly increased in cultured VSMCs stimulated by platelet-derived growth factor-BB (PDGF-BB). A CCK-8 assay, transwell assay, and scratch test were performed to measure the effect of miR-411-5p on the proliferation and migration of PDGF-BB-treated VSMCs. MiR-411-5p promoted expression of dedifferentiated phenotype markers such as osteopontin and tropomyosin 4 in PDGF-BB-treated VSMCs. Using mimics and inhibitors, we identified the target of miR-411-5p in PDGF-BB-treated VSMCs and found that calmodulin-regulated spectrin-associated protein-1 (CAMSAP1) was involved in the phenotypic switch mediated by PDGF-BB.By inhibiting expression of CAMSAP1, miR-411-5p enhanced the proliferation, migration, and phenotype switch of VSMCs.Blockade of miR-411-5p interaction with CAMSAP1 is a promising approach to treat in-stent restenosis.


Subject(s)
Becaplermin , Cell Movement , Cell Proliferation , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Becaplermin/pharmacology , Cells, Cultured , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Rats, Sprague-Dawley , Male , Osteopontin/metabolism , Osteopontin/genetics
13.
Int J Med Sci ; 21(8): 1414-1427, 2024.
Article in English | MEDLINE | ID: mdl-38903916

ABSTRACT

Glutamine (Gln), known as the most abundant free amino acid, is widely spread in human body. In this study, we demonstrated the protective effects of glutamine against mouse abdominal aortic aneurysm (AAA) induced by both angiotensin II (AngII) and calcium phosphate (Ca3(PO4)2) in vivo, which was characterized with lower incidence of mouse AAA. Moreover, histomorphological staining visually presented more intact elastic fiber and less collagen deposition in abdominal aortas of mice treated by glutamine. Further, we found glutamine inhibited the excessive production of reactive oxide species (ROS), activity of matrix metalloproteinase (MMP), M1 macrophage activation, and apoptosis of vascular smooth muscle cells (VSMCs) in suprarenal abdominal aortas of mice, what's more, the high expressions of MMP-2 protein, MMP-9 protein, pro-apoptotic proteins, and IL-6 as well as TNF-α in protein and mRNA levels in cells treated by AngII were down-regulated by glutamine. Collectively, these results revealed that glutamine protected against mouse AAA through inhibiting apoptosis of VSMCs, M1 macrophage activation, oxidative stress, and extracellular matrix degradation.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal , Apoptosis , Glutamine , Macrophage Activation , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Oxidative Stress , Animals , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Aortic Aneurysm, Abdominal/metabolism , Apoptosis/drug effects , Mice , Glutamine/pharmacology , Angiotensin II/pharmacology , Macrophage Activation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/cytology , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Disease Models, Animal , Male , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Aorta, Abdominal/pathology , Aorta, Abdominal/drug effects , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Calcium Phosphates
14.
Aging (Albany NY) ; 16(13): 10749-10764, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38944813

ABSTRACT

Cathepsin L (CTSL) has been implicated in aging and age-related diseases, such as cardiovascular diseases, specifically atherosclerosis. However, the underlying mechanism(s) is not well documented. Recently, we demonstrated a role of CUT-like homeobox 1 (CUX1) in regulating the p16INK4a-dependent cellular senescence in human endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) via its binding to an atherosclerosis-associated functional SNP (fSNP) rs1537371 on the CDKN2A/B locus. In this study, to determine if CTSL, which was reported to proteolytically activate CUX1, regulates cellular senescence via CUX1, we measured the expression of CTSL, together with CUX1 and p16INK4a, in human ECs and VSMCs undergoing senescence. We discovered that CUX1 is not a substrate that is cleaved by CTSL. Instead, CTSL is an upstream regulator that activates CUX1 transcription indirectly in a process that requires the proteolytic activity of CTSL. Our findings suggest that there is a transcription factor in between CTSL and CUX1, and cleavage of this factor by CTSL can activate CUX1 transcription, inducing endothelial senescence. Thus, our findings provide new insights into the signal transduction pathway that leads to atherosclerosis-associated cellular senescence.


Subject(s)
Cathepsin L , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , Homeodomain Proteins , Muscle, Smooth, Vascular , Repressor Proteins , Humans , Cellular Senescence/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cathepsin L/metabolism , Cathepsin L/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Up-Regulation , Endothelial Cells/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Human Umbilical Vein Endothelial Cells , Cells, Cultured
15.
Biomolecules ; 14(6)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38927112

ABSTRACT

De-differentiation and subsequent increased proliferation and inflammation of vascular smooth muscle cells (VSMCs) is one of the mechanisms of atherogenesis. Maintaining VSMCs in a contractile differentiated state is therefore a promising therapeutic strategy for atherosclerosis. We have reported the 18-base myogenetic oligodeoxynucleotide, iSN04, which serves as an anti-nucleolin aptamer and promotes skeletal and myocardial differentiation. The present study investigated the effect of iSN04 on VSMCs because nucleolin has been reported to contribute to VSMC de-differentiation under pathophysiological conditions. Nucleolin is localized in the nucleoplasm and nucleoli of both rat and human VSMCs. iSN04 without a carrier was spontaneously incorporated into VSMCs, indicating that iSN04 would serve as an anti-nucleolin aptamer. iSN04 treatment decreased the ratio of 5-ethynyl-2'-deoxyuridine (EdU)-positive proliferating VSMCs and increased the expression of α-smooth muscle actin, a contractile marker of VSMCs. iSN04 also suppressed angiogenesis of mouse aortic rings ex vivo, which is a model of pathological angiogenesis involved in plaque formation, growth, and rupture. These results demonstrate that antagonizing nucleolin with iSN04 preserves VSMC differentiation, providing a nucleic acid drug candidate for the treatment of vascular disease.


Subject(s)
Aptamers, Nucleotide , Cell Differentiation , Cell Proliferation , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Nucleolin , Phosphoproteins , RNA-Binding Proteins , Animals , RNA-Binding Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Aptamers, Nucleotide/pharmacology , Cell Proliferation/drug effects , Phosphoproteins/metabolism , Cell Differentiation/drug effects , Humans , Rats , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Mice , Cells, Cultured , Oligodeoxyribonucleotides/pharmacology , Male , Rats, Sprague-Dawley , Mice, Inbred C57BL
16.
Exp Cell Res ; 440(2): 114136, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38909881

ABSTRACT

Considering the importance of alternative methodologies to animal experimentation, we propose an organoid-based biological model for in vitro blood vessel generation, achieved through co-culturing endothelial and vascular smooth muscle cells (VSMCs). Initially, the organoids underwent comprehensive characterization, revealing VSMCs (α-SMA + cells) at the periphery and endothelial cells (CD31+ cells) at the core. Additionally, ephrin B2 and ephrin B4, genes implicated in arterial and venous formation respectively, were used to validate the obtained organoid. Moreover, the data indicates exclusive HIF-1α expression in VSMCs, identified through various methodologies. Subsequently, we tested the hypothesis that the generated blood vessels have the capacity to modulate the osteogenic phenotype, demonstrating the ability of HIF-1α to promote osteogenic signals, primarily by influencing Runx2 expression. Overall, this study underscores that the methodology employed to create blood vessel organoids establishes an experimental framework capable of producing a 3D culture model of both venous and arterial endothelial tissues. This model effectively guides morphogenesis from mesenchymal stem cells through paracrine signaling, ultimately leading to an osteogenic acquisition phenotype, with the dynamic involvement of HIF-1α.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Organoids , Osteogenesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Osteogenesis/genetics , Organoids/metabolism , Organoids/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Cells, Cultured , Blood Vessels/metabolism , Blood Vessels/cytology , Blood Vessels/growth & development , Coculture Techniques/methods , Cell Differentiation , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology
17.
Cell Signal ; 121: 111276, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936786

ABSTRACT

(-)-Epicatechin gallate (ECG) is beneficial to the treatment of cardiovascular diseases (CVDs), especially atherosclerosis (AS) through antioxidant stress, but there is a lack of detailed mechanism research. In this study, the therapeutic target of ECG was determined by crossing the drug target and disease target of CVDs and AS. The combination ability of ECG with important targets was verified by Discovery Studio software. The abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by Ang-II and the oxidative damage of AML 12 induced by H2O2 were established to verify the reliability of ECG intervention on the target protein. A total of 120 ECG targets for the treatment of CVDs-AS were predicted by network pharmacology. The results of molecular docking showed that ECG has strong binding force with VEGFA, MMP-9, CASP3 and MMP-2 domains. In vitro experiments confirmed that ECG significantly reduced the expression of VEGFA, MMP-9, CASP3 and MMP-2 in Ang-II-induced VSMCs, and also blocked the abnormal proliferation, oxidative stress and inflammatory reaction of VSMCs by inhibiting the phosphorylation of PI3K signaling pathway. At the same time, ECG also interfered with H2O2-induced oxidative damage of AML 12 cells, decreased the expression of ROS and MDA and cell foaming, and increased the activities of antioxidant enzymes such as SOD, thus playing a protective role.


Subject(s)
Atherosclerosis , Catechin , Cell Proliferation , Hydrogen Peroxide , Molecular Docking Simulation , Muscle, Smooth, Vascular , Oxidative Stress , Oxidative Stress/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Proliferation/drug effects , Atherosclerosis/metabolism , Atherosclerosis/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/cytology , Hydrogen Peroxide/pharmacology , Humans , Signal Transduction/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Cell Line , Vascular Endothelial Growth Factor A/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Angiotensin II/pharmacology , Matrix Metalloproteinase 9/metabolism , Antioxidants/pharmacology
19.
Redox Biol ; 73: 103203, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823208

ABSTRACT

Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2's specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease's activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.


Subject(s)
Endopeptidase Clp , Mitochondria , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , Sirtuin 1 , Animals , Humans , Mice , Cell Differentiation , Endopeptidase Clp/metabolism , Endopeptidase Clp/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics
20.
Biochem Biophys Res Commun ; 725: 150248, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38870847

ABSTRACT

The excessive migration and proliferation of vascular smooth muscle cells (VSMCs) plays a vital role in vascular intimal hyperplasia. CIRBP is involved in the proliferation of various cancer cells. This study was aimed to explore the role of CIRBP in the proliferation and migration of VSMCs. Adenovirus was used to interfere with cold-inducible RNA-binding protein (CIRBP) expression, while lentivirus was used to overexpress Ras homolog enriched in brain (Rheb). Western blotting and qRT-PCR were used to evaluate the expression of CIRBP, Rheb, and mechanistic target of rapamycin complex 1 (mTORC1) activity. The cell proliferation was determined by Ki67 immunofluorescence staining and CCK-8 assay. The wound healing assay was performed to assess cell migration. Additionally, immunohistochemistry was conducted to explore the role of CIRBP in intimal hyperplasia after vascular injury. We found that silencing CIRBP inhibited the proliferation and migration of VSMCs, decreased the expression of Rheb and mTORC1 activity. Restoration of mTORC1 activity via insulin or overexpression of Rheb via lentiviral transfection both attenuated the inhibitory effects of silencing CIRBP on the proliferation and migration of VSMCs. Moreover, Rheb overexpression abolished the inhibitory effect of silencing CIRBP on mTORC1 activity in VSMCs. CIRBP was upregulated in the injured carotid artery. Silencing CIRBP ameliorated intimal hyperplasia after vascular injury. In the summary, silencing CIRBP attenuates mTORC1 activity via reducing Rheb expression, thereby supressing the proliferation and migration of VSMCs and intimal hyperplasia after vascular injury.


Subject(s)
Cell Movement , Cell Proliferation , Mechanistic Target of Rapamycin Complex 1 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , RNA-Binding Proteins , Ras Homolog Enriched in Brain Protein , Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Ras Homolog Enriched in Brain Protein/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/cytology , Cells, Cultured , Signal Transduction , Male , Rats , Rats, Sprague-Dawley , Humans
SELECTION OF CITATIONS
SEARCH DETAIL