Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 462
Filter
1.
Development ; 148(7)2021 03 31.
Article in English | MEDLINE | ID: mdl-33789914

ABSTRACT

Smooth muscle cells (SMCs) represent a major structural and functional component of many organs during embryonic development and adulthood. These cells are a crucial component of vertebrate structure and physiology, and an updated overview of the developmental and functional process of smooth muscle during organogenesis is desirable. Here, we describe the developmental origin of SMCs within different tissues by comparing their specification and differentiation with other organs, including the cardiovascular, respiratory and intestinal systems. We then discuss the instructive roles of smooth muscle in the development of such organs through signaling and mechanical feedback mechanisms. By understanding SMC development, we hope to advance therapeutic approaches related to tissue regeneration and other smooth muscle-related diseases.


Subject(s)
Embryonic Development , Muscle, Smooth/growth & development , Myocytes, Smooth Muscle/physiology , Vertebrates/growth & development , Animals , Animals, Genetically Modified , Cardiovascular System , Cell Differentiation/physiology , Gastrointestinal Tract , Lung , Mesoderm , Muscle, Smooth/cytology , Muscle, Smooth/embryology , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/cytology , Organogenesis/physiology , Respiratory System , Vertebrates/embryology
2.
Circ Res ; 125(11): 1006-1018, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31590613

ABSTRACT

RATIONALE: Elastin is an important ECM (extracellular matrix) protein in large and small arteries. Vascular smooth muscle cells (SMCs) produce the layered elastic laminae found in elastic arteries but synthesize little elastin in muscular arteries. However, muscular arteries have a well-defined internal elastic lamina (IEL) that separates endothelial cells (ECs) from SMCs. The extent to which ECs contribute elastin to the IEL is unknown. OBJECTIVE: To use targeted elastin (Eln) deletion in mice to explore the relative contributions of SMCs and ECs to elastic laminae formation in different arteries. METHODS AND RESULTS: We used SMC- and EC-specific Cre recombinase transgenes with a novel floxed Eln allele to focus gene inactivation in mice. Inactivation of Eln in SMCs using Sm22aCre resulted in depletion of elastic laminae in the arterial wall with the exception of the IEL and SMC clusters in the outer media near the adventitia. Inactivation of elastin in ECs using Tie2Cre or Cdh5Cre resulted in normal medial elastin and a typical IEL in elastic arteries. In contrast, the IEL was absent or severely disrupted in muscular arteries. Interruptions in the IEL resulted in neointimal formation in the ascending aorta but not in muscular arteries. CONCLUSIONS: Combined with lineage-specific fate mapping systems, our knockout results document an unexpected heterogeneity in vascular cells that produce the elastic laminae. SMCs and ECs can independently form an IEL in most elastic arteries, whereas ECs are the major source of elastin for the IEL in muscular and resistance arteries. Neointimal formation at IEL disruptions in the ascending aorta confirms that the IEL is a critical physical barrier between SMCs and ECs in the large elastic arteries. Our studies provide new information about how SMCs and ECs contribute elastin to the arterial wall and how local elastic laminae defects may contribute to cardiovascular disease.


Subject(s)
Elastic Tissue/metabolism , Elastin/metabolism , Endothelial Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Arteries/growth & development , Arteries/metabolism , Blood Pressure , Cell Lineage , Cell Proliferation , Elastic Tissue/growth & development , Elastic Tissue/ultrastructure , Elastin/deficiency , Elastin/genetics , Endothelial Cells/ultrastructure , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Neointima , Signal Transduction
3.
J Cell Mol Med ; 24(8): 4762-4772, 2020 04.
Article in English | MEDLINE | ID: mdl-32155686

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation is the pathological base of vascular remodelling diseases. Circular RNAs (circRNAs) are important regulators involved in various biological processes. However, the function of circRNAs in VSMC proliferation regulation remains largely unknown. This study was conducted to identify the key differentially expressed circRNAs (DEcircRNAs) and predict their functions in human aortic smooth muscle cell (HASMC) proliferation. To achieve this, DEcircRNAs between proliferative and quiescent HASMCs were detected using a microarray, followed by quantitative real-time RT-PCR validation. A DEcircRNA-miRNA-DEmRNA network was constructed, and functional annotation was performed using Gene Ontology (GO) and KEGG pathway analysis. The function of hsa_circ_0002579 in HASMC proliferation was analysed by Western blot. The functional annotation of the DEcircRNA-miRNA-DEmRNA network indicated that the four DEcircRNAs might play roles in the TGF-ß receptor signalling pathway, Ras signalling pathway, AMPK signalling pathway and Wnt signalling pathway. Twenty-seven DEcircRNAs with coding potential were screened. Hsa_circ_0002579 might be a pro-proliferation factor of HASMC. Overall, our study identified the key DEcircRNAs between proliferative and quiescent HASMCs, which might provide new important clues for exploring the functions of circRNAs in vascular remodelling diseases.


Subject(s)
Aorta/growth & development , Cell Proliferation/genetics , Muscle, Smooth, Vascular/growth & development , RNA, Circular/genetics , Aorta/metabolism , Cell Culture Techniques , Gene Expression Regulation, Developmental/genetics , Gene Ontology , Gene Regulatory Networks/genetics , Humans , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
4.
Am J Physiol Heart Circ Physiol ; 318(4): H976-H984, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32142377

ABSTRACT

There is a sustained reduction in arterial blood pressure that occurs in aged adults following exposure to acute leg heating. We tested the hypothesis that acute leg heating would decrease arterial blood pressure in aged adults secondary to sympathoinhibition. We exposed 13 young and 10 aged adults to 45 min of leg heating. Muscle sympathetic nerve activity (radial nerve) was measured before leg heating (preheat) and 30 min after (recovery) and is expressed as burst frequency. Neurovascular transduction was examined by assessing the slope of the relation between muscle sympathetic nerve activity and leg vascular conductance measured at rest and during isometric handgrip exercise performed to fatigue. Arterial blood pressure was well maintained in young adults (preheat, 86 ± 6 mmHg vs. recovery, 88 ± 7 mmHg; P = 0.4) due to increased sympathetic nerve activity (preheat, 16 ± 7 bursts/min vs. recovery, 22 ± 10 bursts/min; P < 0.01). However, in aged adults, sympathetic nerve activity did not differ from preheat (37 ± 5 bursts/min) to recovery (33 ± 6 bursts/min, P = 0.1), despite a marked reduction in arterial blood pressure (preheat, 101 ± 7 mmHg vs. recovery, 94 ± 6 mmHg; P < 0.01). Neurovascular transduction did not differ from preheat to recovery for either age group (P ≥ 0.1). The reduction in arterial blood pressure that occurs in aged adults following exposure to acute leg heating is mediated, in part, by a sympathoinhibitory effect that alters the compensatory neural response to hypotension.NEW & NOTEWORTHY There is a sustained reduction in arterial blood pressure that occurs in aged adults following exposure to acute leg heating. However, the neurovascular mechanisms mediating this response remain unknown. Our findings demonstrate for the first time that this reduction in arterial blood pressure is mediated, in part, by a sympathoinhibitory effect that alters the compensatory neural response to hypotension in aged adults.


Subject(s)
Aging/physiology , Blood Pressure , Heat-Shock Response , Sympathetic Nervous System/physiology , Adult , Aged , Female , Hand Strength , Humans , Leg/growth & development , Leg/physiology , Male , Middle Aged , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology , Neural Conduction , Sympathetic Nervous System/growth & development
5.
Clin Sci (Lond) ; 134(15): 2019-2035, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32706027

ABSTRACT

OBJECTIVE: Transient receptor potential (TRP) melastatin 7 (TRPM7) cation channel, a dual-function ion channel/protein kinase, regulates vascular smooth muscle cell (VSMC) Mg2+ homeostasis and mitogenic signaling. Mechanisms regulating vascular growth effects of TRPM7 are unclear, but epidermal growth factor (EGF) may be important because it is a magnesiotropic hormone involved in cellular Mg2+ regulation and VSMC proliferation. Here we sought to determine whether TRPM7 is a downstream target of EGF in VSMCs and if EGF receptor (EGFR) through TRPM7 influences VSMC function. Approach and results: Studies were performed in primary culture VSMCs from rats and humans and vascular tissue from mice deficient in TRPM7 (TRPM7+/Δkinase and TRPM7R/R). EGF increased expression and phosphorylation of TRPM7 and stimulated Mg2+ influx in VSMCs, responses that were attenuated by gefitinib (EGFR inhibitor) and NS8593 (TRPM7 inhibitor). Co-immunoprecipitation (IP) studies, proximity ligation assay (PLA) and live-cell imaging demonstrated interaction of EGFR and TRPM7, which was enhanced by EGF. PP2 (c-Src inhibitor) decreased EGF-induced TRPM7 activation and prevented EGFR-TRPM7 association. EGF-stimulated migration and proliferation of VSMCs were inhibited by gefitinib, PP2, NS8593 and PD98059 (ERK1/2 inhibitor). Phosphorylation of EGFR and ERK1/2 was reduced in VSMCs from TRPM7+/Δkinase mice, which exhibited reduced aortic wall thickness and decreased expression of PCNA and Notch 3, findings recapitulated in TRPM7R/R mice. CONCLUSIONS: We show that EGFR directly interacts with TRPM7 through c-Src-dependent processes. Functionally these phenomena regulate [Mg2+]i homeostasis, ERK1/2 signaling and VSMC function. Our findings define a novel signaling cascade linking EGF/EGFR and TRPM7, important in vascular homeostasis.


Subject(s)
Epidermal Growth Factor/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Animals , CSK Tyrosine-Protein Kinase/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Magnesium/metabolism , Mice, Inbred C57BL , Morphogenesis , Muscle, Smooth, Vascular/growth & development , Phosphorylation , Primary Cell Culture , Rats, Inbred WKY
6.
Exp Cell Res ; 376(1): 77-85, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30690026

ABSTRACT

Cyclic stretch regulates proliferation of vascular smooth muscle cells (VSMCs) during hypertension-induced vascular remodeling, but the underlying mechanisms remain to be studied. Connective tissue growth factor (CTGF) has been reported associated with several cellular function such as proliferation,migration and adhesion. Herein, the role of CTGF in VSMCs was investigated in response to mechanical cyclic stretch. Here we show that CTGF is up-regulated both in vivo and in vitro during hypertension. Overexpression of CTGF markedly promoted VSMC proliferation, whereas CTGF knockdown attenuated cyclic stretch-induced proliferation. Furthermore, 3'UTR reporter assays revealed that microRNA-19b-3p (miR-19b-3p) directly regulates CTGF expression. Under pathological condition (e.g. 15% cyclic stretch), miR-19b-3p expression was significantly down-regulated; conversely miR-19b-3p overexpression blocked VSMC proliferation. Taken together, these findings indicate that pathological cyclic stretch induces vascular remodeling by promoting VSMC proliferation via miR-19b-3p/CTGF pathway, and point to CTGF as a potential therapeutic target for hypertension.


Subject(s)
Cell Proliferation/genetics , Connective Tissue Growth Factor/genetics , Hypertension/genetics , MicroRNAs/genetics , Muscle, Smooth, Vascular/growth & development , 3' Untranslated Regions/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Hypertension/drug therapy , Hypertension/pathology , Muscle, Smooth, Vascular/metabolism , Signal Transduction/genetics
7.
Am J Respir Crit Care Med ; 200(5): 617-627, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30817168

ABSTRACT

Rationale: Glycolytic shift is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). It remains unknown how glycolysis is increased and how increased glycolysis contributes to pulmonary vascular remodeling in PAH.Objectives: To determine whether increased glycolysis is caused by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and how PFKFB3-driven glycolysis induces vascular remodeling in PAH.Methods: PFKFB3 levels were measured in pulmonary arteries of patients and animals with PAH. Lactate levels were assessed in lungs of animals with PAH and in pulmonary artery smooth muscle cells (PASMCs). Genetic and pharmacologic approaches were used to investigate the role of PFKFB3 in PAH.Measurements and Main Results: Lactate production was elevated in lungs of PAH rodents and in platelet-derived growth factor-treated PASMCs. PFKFB3 protein was higher in pulmonary arteries of patients and rodents with PAH, in PASMCs of patients with PAH, and in platelet-derived growth factor-treated PASMCs. PFKFB3 inhibition by genetic disruption and chemical inhibitor attenuated phosphorylation/activation of extracellular signal-regulated kinase (ERK1/2) and calpain-2, and vascular remodeling in PAH rodent models, and reduced platelet-derived growth factor-induced phosphorylation/activation of ERK1/2 and calpain-2, collagen synthesis and proliferation of PASMCs. ERK1/2 inhibition attenuated phosphorylation/activation of calpain-2, and vascular remodeling in Sugen/hypoxia PAH rats, and reduced lactate-induced phosphorylation/activation of calpain-2, collagen synthesis, and proliferation of PASMCs. Calpain-2 inhibition reduced lactate-induced collagen synthesis and proliferation of PASMCs.Conclusions: Upregulated PFKFB3 mediates collagen synthesis and proliferation of PASMCs, contributing to vascular remodeling in PAH. The mechanism is through the elevation of glycolysis and lactate that results in the activation of calpain by ERK1/2-dependent phosphorylation of calpain-2.


Subject(s)
Cell Proliferation/drug effects , Muscle, Smooth, Vascular/growth & development , Phosphofructokinase-2/blood , Phosphofructokinase-2/metabolism , Pulmonary Arterial Hypertension/blood , Pulmonary Arterial Hypertension/physiopathology , Vascular Remodeling/physiology , Animals , Disease Models, Animal , Humans , Male , Mice , Rats
8.
Int J Mol Sci ; 21(24)2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33322781

ABSTRACT

Amine-coated biodegradable materials based on synthetic polymers have a great potential for tissue remodeling and regeneration because of their excellent processability and bioactivity. In the present study, we have investigated the influence of various chemical compositions of amine plasma polymer (PP) coatings and the influence of the substrate morphology, represented by polystyrene culture dishes and polycaprolactone nanofibers (PCL NFs), on the behavior of vascular smooth muscle cells (VSMCs). Although all amine-PP coatings improved the initial adhesion of VSMCs, 7-day long cultivation revealed a clear preference for the coating containing about 15 at.% of nitrogen (CPA-33). The CPA-33 coating demonstrated the ideal combination of good water stability, a sufficient amine group content, and favorable surface wettability and morphology. The nanostructured morphology of amine-PP-coated PCL NFs successfully slowed the proliferation rate of VSMCs, which is essential in preventing restenosis of vascular replacements in vivo. At the same time, CPA-33-coated PCL NFs supported the continuous proliferation of VSMCs during 7-day long cultivation, with no significant increase in cytokine secretion by RAW 264.7 macrophages. The CPA-33 coating deposited on biodegradable PCL NFs therefore seems to be a promising material for manufacturing small-diameter vascular grafts, which are still lacking on the current market.


Subject(s)
Amines/chemistry , Coated Materials, Biocompatible/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nanofibers/chemistry , Plasma/chemistry , Polymers/chemistry , Amines/adverse effects , Amines/immunology , Amines/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Proliferation/drug effects , Cells, Cultured , Coated Materials, Biocompatible/adverse effects , Coated Materials, Biocompatible/chemistry , Macrophages/drug effects , Macrophages/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/metabolism , Nanofibers/adverse effects , Photoelectron Spectroscopy , Plasma/immunology , Polyesters/chemistry , Polymers/adverse effects , Polymers/pharmacology , RAW 264.7 Cells , Rats , Surface Properties/drug effects , Tissue Scaffolds/adverse effects , Tissue Scaffolds/chemistry
9.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L999-L1012, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30908936

ABSTRACT

Lung diseases with hypoxia are complicated by pulmonary hypertension, leading to heart failure and death. No pharmacological treatment exists. Increased proinflammatory cytokines are found in hypoxic patients, suggesting an inflammatory pathogenesis. Caspase-1, the effector of the inflammasome, mediates inflammation through activation of the proinflammatory cytokines interleukin (IL)-18 and IL-1ß. Here, we investigate inflammasome-related mechanisms that can trigger hypoxia-induced pulmonary hypertension. Our aim was to examine whether caspase-1 induces development of hypoxia-related pulmonary hypertension and is a suitable target for therapy. Wild-type (WT) and caspase-1-/- mice were exposed to 10% oxygen for 14 days. Hypoxic caspase-1-/- mice showed lower pressure and reduced muscularization in pulmonary arteries, as well as reduced right ventricular remodeling compared with WT. Smooth muscle cell (SMC) proliferation was reduced in caspase-1-deficient pulmonary arteries and in WT arteries treated with a caspase-1 inhibitor. Impaired inflammation was shown in hypoxic caspase-1-/- mice by abolished pulmonary influx of immune cells and lower levels of IL-18, IL-1ß, and IL-6, which were also reduced in the medium surrounding caspase-1 abrogated pulmonary arteries. By adding IL-18 or IL-1ß to caspase-1-deficient pulmonary arteries, SMC proliferation was retained. Furthermore, inhibition of both IL-6 and phosphorylated STAT3 reduced proliferation of SMC in vitro, indicating IL-18, IL-6, and STAT3 as downstream mediators of caspase-1-induced SMC proliferation in pulmonary arteries. Caspase-1 induces SMC proliferation in pulmonary arteries through the caspase-1/IL-18/IL-6/STAT3 pathway, leading to pulmonary hypertension in mice exposed to hypoxia. We propose that caspase-1 inhibition is a potential target for treatment of pulmonary hypertension.


Subject(s)
Caspase 1/genetics , Cell Hypoxia/physiology , Hypertension, Pulmonary/pathology , Myocytes, Smooth Muscle/physiology , Ventricular Function, Right/physiology , Animals , Cell Line , Cell Proliferation/genetics , Humans , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/growth & development , Pulmonary Artery/cytology , Pulmonary Artery/pathology , STAT3 Transcription Factor/metabolism
10.
IUBMB Life ; 71(1): 125-133, 2019 01.
Article in English | MEDLINE | ID: mdl-30291803

ABSTRACT

The long-term failure of vein grafts due to neointimal hyperplasia remains a difficult problem in cardiovascular surgery. Exploring novel approaches to prevent neointimal hyperplasia is important. MicroRNA-146a (miR-146a) plays an essential role in promoting vascular smooth muscle cell (VSMC) proliferation. Thus, the aim of the present study is to investigate whether adenovirus-mediated miR-146a sponge (Ad-miR-146a-SP) gene therapy could attenuate neointimal formation in rat vein grafts. (Ad-miR-146a-SP) was constructed to transfect cultured VSMCs and grafted veins. To improve the efficiency of transferring the miR-146a sponge gene into the grafted veins, 20% poloxamer F-127 gel incorporated with 0.25% trypsin was used to increase adenovirus contact time and penetration. miR-146a-SP transduction significantly reduced the expression of miR-146a both in cultured VSMCs and vein grafts. miR-146a sponge markedly attenuated VSMC proliferation and migration. Consistent with this, miR-146a sponge gene therapy significantly attenuated neointimal formation and also improved blood flow in the vein grafts. Mechanistically, we identified the Krüppel-like factor 4(KLF4) as a potential downstream target gene of miR-146a in vein grafts. Our data show that miR-146a sponge gene therapy could effectively reduce miR-146a activity and attenuate neointimal formation in vein grafts, suggesting its potential therapeutic application for prevention of vein graft failure. © 2018 IUBMB Life, 71(1):125-133, 2019.


Subject(s)
Genetic Therapy , MicroRNAs/genetics , Neointima/therapy , Veins/growth & development , Adenoviridae/genetics , Animals , Blood Vessel Prosthesis , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , MicroRNAs/pharmacology , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Rats , Veins/physiopathology
11.
J Anat ; 234(4): 465-479, 2019 04.
Article in English | MEDLINE | ID: mdl-30793310

ABSTRACT

Teeth are richly supported by blood vessels and peripheral nerves. The aim of this study was to describe in detail the developmental time-course and localization of blood vessels during early tooth formation and to compare that to innervation, as well as to address the putative role of vascular endothelial growth factor (VEGF), which is an essential regulator of vasculature development, in this process. The localization of blood vessels and neurites was compared using double immunofluorescence staining on sections at consecutive stages of the embryonic (E) and postnatal (PN) mandibular first molar tooth germ (E11-PN7). Cellular mRNA expression domains of VEGF and its signaling receptor VEGFR2 were studied using sectional radioactive in situ hybridization. Expression of VEGF mRNA and the encoded protein were studied by RT-PCR and western blot analysis, respectively, in the cap and early bell stage tooth germs, respectively. VEGFR2 was immunolocalized on tooth tissue sections. Smooth muscle cells were investigated by anti-alpha smooth muscle actin (αSMA) antibodies. VEGF showed developmentally regulated epithelial and mesenchymal mRNA expression domains including the enamel knot signaling centers that correlated with the growth and navigation of the blood vessels expressing Vegfr2 and VEGFR2 to the dental papilla and enamel organ. Developing blood vessels were present in the jaw mesenchyme including the presumptive dental mesenchyme before the appearance of the epithelial dental placode and dental neurites. Similarly, formation of a blood vessel plexus around the bud stage tooth germ and ingrowth of vessels into dental papilla at E14 preceded ingrowth of neurites. Subsequently, pioneer blood vessels in the dental papilla started to receive smooth muscle coverage at the early embryonic bell stage. Establishment and patterning of the blood vessels and nerves during tooth formation are developmentally regulated, stepwise processes that likely involve differential patterning mechanisms. Development of tooth vascular supply is proposed to be regulated by local, tooth-specific regulation by epithelial-mesenchymal tissue interactions and involving tooth target expressed VEGF signaling. Further investigations on tooth vascular development by local VEGF signaling, as well as how tooth innervation and development of blood vessels are integrated with advancing tooth organ formation by local signaling mechanisms, are warranted.


Subject(s)
Blood Vessels/growth & development , Tooth , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Body Patterning , Gene Expression Regulation, Developmental , Mice , Muscle, Smooth, Vascular/growth & development , Odontogenesis , Signal Transduction , Tooth/embryology , Tooth/innervation , Tooth/metabolism , Tooth Germ/embryology , Tooth Germ/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Am J Physiol Cell Physiol ; 315(6): C850-C862, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30257103

ABSTRACT

Vascular smooth muscle cell (VSMC) migration contributes to neointimal formation after vascular injury. We previously demonstrated that copper (Cu) transporter ATP7A is involved in platelet-derived growth factor (PDGF)-induced VSMC migration in a Cu- and Rac1-dependent manner. The underlying mechanism is still unknown. Here we show that ATP7A interacts with IQGAP1, a Rac1 and receptor tyrosine kinase binding scaffolding proteins, which mediates PDGF-induced VSMC migration and vascular remodeling. In cultured rat aortic SMCs, PDGF stimulation rapidly promoted ATP7A association with IQGAP1 and Rac1 and their translocation to the lipid rafts and leading edge. Cotransfection assay revealed that ATP7A directly bound to NH2-terminal domain of IQGAP1. Functionally, either ATP7A or IQGAP1 depletion using siRNA significantly inhibited PDGF-induced VSMC migration without additive effects, suggesting that IQGAP1 and ATP7A are in the same axis to promote migration. Furthermore, IQGAP1 siRNA blocked PDGF-induced ATP7A association with Rac1 as well as its translocation to leading edge, while PDGF-induced IQGAP1 translocation was not affected by ATP7A siRNA or Cu chelator. Overexpression of mutant IQGAP1 lacking a Rac1 binding site prevented PDGF-induced translocation of Rac1, but not ATP7A, to the leading edge, thereby inhibiting lamellipodia formation and VSMC migration. In vivo, ATP7A colocalized with IQGAP1 at neointimal VSMCs in a mice wire injury model, while neointimal formation and extracellular matrix deposition induced by vascular injury were inhibited in ATP7A mutant mice with reduced Cu transporter function. In summary, IQGAP1 functions as ATP7A and Rac1 binding scaffolding protein to organize PDGF-dependent ATP7A translocation to the lamellipodial leading edge, thereby promoting VSMC migration and vascular remodeling.


Subject(s)
Copper-Transporting ATPases/genetics , Platelet-Derived Growth Factor/genetics , Vascular Remodeling/genetics , rac1 GTP-Binding Protein/genetics , ras GTPase-Activating Proteins/genetics , Animals , Aorta/cytology , Aorta/metabolism , Cell Movement/genetics , Copper/chemistry , Copper/metabolism , Gene Expression Regulation, Developmental/genetics , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Mice , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Phosphorylation , Protein Binding , Rats
13.
Arterioscler Thromb Vasc Biol ; 37(5): 930-941, 2017 05.
Article in English | MEDLINE | ID: mdl-28254817

ABSTRACT

OBJECTIVE: Williams syndrome is characterized by obstructive aortopathy attributable to heterozygous loss of ELN, the gene encoding elastin. Lesions are thought to result primarily from excessive smooth muscle cell (SMC) proliferation and consequent medial expansion, although an initially smaller caliber and increased stiffness of the aorta may contribute to luminal narrowing. The relative contributions of such abnormalities to the obstructive phenotype had not been defined. APPROACH AND RESULTS: We quantified determinants of luminal stenosis in thoracic aortas of Eln-/- mice incompletely rescued by human ELN. Moderate obstruction was largely because of deficient circumferential growth, most prominently of ascending segments, despite increased axial growth. Medial thickening was evident in these smaller diameter elastin-deficient aortas, with medial area similar to that of larger diameter control aortas. There was no difference in cross-sectional SMC number between mutant and wild-type genotypes at multiple stages of postnatal development. Decreased elastin content was associated with medial fibrosis and reduced aortic distensibility because of increased structural stiffness but preserved material stiffness. Elastin-deficient SMCs exhibited greater contractile-to-proliferative phenotypic modulation in vitro than in vivo. We confirmed increased medial collagen without evidence of increased medial area or SMC number in a small ascending aorta with thickened media of a Williams syndrome subject. CONCLUSIONS: Deficient circumferential growth is the predominant mechanism for moderate obstructive aortic disease resulting from partial elastin deficiency. Our findings suggest that diverse aortic manifestations in Williams syndrome result from graded elastin content, and SMC hyperplasia causing medial expansion requires additional elastin loss superimposed on ELN haploinsufficiency.


Subject(s)
Aorta, Thoracic/growth & development , Aortic Diseases/physiopathology , Elastin/metabolism , Williams Syndrome/physiopathology , Adult , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Constriction, Pathologic , Disease Models, Animal , Elastin/deficiency , Elastin/genetics , Fibrosis , Genetic Predisposition to Disease , Humans , Hyperplasia , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Time Factors , Vascular Stiffness , Vasoconstriction , Williams Syndrome/genetics , Williams Syndrome/metabolism , Williams Syndrome/pathology
14.
Sheng Li Xue Bao ; 70(5): 548-556, 2018 Oct 25.
Article in Zh | MEDLINE | ID: mdl-30377694

ABSTRACT

The vertebrate vascular system development is a very important and complicated process. MicroRNAs regulate gene expression at transcriptional and post-transcriptional levels and play important roles in many physiological and pathological processes. MicroRNAs mainly participate in the regulation of vascular smooth muscle cell and vascular endothelial cell development. In this paper, we summarize the recent progress regarding the microRNAs involved in the vascular development. In particular, we focus on the microRNAs including miR-126, miR-17/92 family in endothelial cell's regulation, and miR-143/145 family, miR-21 in vascular smooth muscle cell's regulation. The future research on the role of microRNAs in vascular development is also prospected.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation , MicroRNAs/genetics , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/cytology , Animals , Cell Differentiation , Humans , Muscle, Smooth, Vascular/cytology
15.
Am J Physiol Heart Circ Physiol ; 312(4): H832-H841, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28159810

ABSTRACT

Systemic hypoxia is a physiological and pathophysiological stress that activates the sympathoadrenal system and, in young adults, leads to peripheral vasodilation. We tested the hypothesis that peripheral vasodilation to graded systemic hypoxia is impaired in older healthy adults and that this age-associated impairment is due to attenuated ß-adrenergic mediated vasodilation and elevated α-adrenergic vasoconstriction. Forearm blood flow was measured (Doppler ultrasound), and vascular conductance (FVC) was calculated in 12 young (24 ± 1 yr) and 10 older (63 ± 2 yr) adults to determine the local dilatory responses to graded hypoxia (90, 85, and 80% O2 saturations) in control conditions, following local intra-arterial blockade of ß-receptors (propranolol), and combined blockade of α- and ß-receptors (phentolamine + propranolol). Under control conditions, older adults exhibited impaired vasodilation to hypoxia compared with young participants at all levels of hypoxia (peak ΔFVC at 80% [Formula: see text] = 4 ± 6 vs. 35 ± 8%; P < 0.01). During ß-blockade, older adults actively constricted at 85 and 80% [Formula: see text] (peak ΔFVC at 80% [Formula: see text] = -13 ± 6%; P < 0.05 vs. control), whereas the response in the young was not significantly impacted (peak ΔFVC = 28 ± 8%). Combined α- and ß-blockade increased the dilatory response to hypoxia in young adults; however, older adults failed to significantly vasodilate (peak ΔFVC at 80% [Formula: see text]= 12 ± 11% vs. 58 ± 11%; P < 0.05). Our findings indicate that peripheral vasodilation to graded systemic hypoxia is significantly impaired in older adults, which cannot be fully explained by altered sympathoadrenal control of vascular tone. Thus, the impairment in hypoxic vasodilation is likely due to attenuated local vasodilatory and/or augmented vasoconstrictor signaling with age.NEW & NOTEWORTHY We found that the lack of peripheral vasodilation during graded systemic hypoxia with aging is not mediated by the sympathoadrenal system, strongly implicating local vascular control mechanisms in this impairment. Understanding these mechanisms may lead to therapeutic advances for improving tissue blood flow and oxygen delivery in aging and disease.


Subject(s)
Hypoxia/physiopathology , Sympathetic Nervous System/physiology , Vasodilation/physiology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Adult , Aged , Aged, 80 and over , Aging/physiology , Blood Gas Analysis , Body Composition , Catecholamines/blood , Female , Forearm/blood supply , Forearm/diagnostic imaging , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology , Regional Blood Flow/physiology , Sympathetic Nervous System/drug effects , Vasodilation/drug effects , Young Adult
16.
Circ Res ; 116(3): 480-8, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25634971

ABSTRACT

There is increasing interest in the role of autophagic flux in maintaining normal vessel wall biology and a growing suspicion that autophagic dysregulation may be a common pathway through which vascular aging and associated pathologies develop. Within endothelial and smooth muscle cells, diverse but important triggers that range from oxidized lipids to ß-amyloid seem to stimulate autophagosome formation potently. In addition, emerging evidence links autophagy to a wide array of vascular processes ranging from angiogenesis to calcification of the vessel wall. Alterations in autophagic flux are also increasingly being implicated in disease processes that include both atherosclerosis and pulmonary hypertension. Finally, recent insights point toward an important role of autophagy in the paracrine regulation of vasoactive substances from the endothelium. Here, we review the progress in understanding how autophagy can contribute to vascular biology and the emerging strategies to target this process for therapeutic benefit.


Subject(s)
Autophagy , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Aging , Amyloid beta-Peptides/metabolism , Animals , Endothelium, Vascular/growth & development , Endothelium, Vascular/physiology , Humans , Lipid Peroxides/metabolism , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology
17.
J Physiol ; 594(8): 2115-24, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26174609

ABSTRACT

Atherosclerosis is a disease of ageing in that its incidence and prevalence increase with age. However, atherosclerosis is also associated with biological ageing, manifest by a number of typical hallmarks of ageing in the atherosclerotic plaque. Thus, accelerated biological ageing may be superimposed on the effects of chronological ageing in atherosclerosis. Tissue ageing is seen in all cells that comprise the plaque, but particularly in vascular smooth muscle cells (VSMCs). Hallmarks of ageing include evidence of cell senescence, DNA damage (including telomere attrition), mitochondrial dysfunction, a pro-inflammatory secretory phenotype, defects in proteostasis, epigenetic changes, deregulated nutrient sensing, and exhaustion of progenitor cells. In this model, initial damage to DNA (genomic, telomeric, mitochondrial and epigenetic changes) results in a number of cellular responses (cellular senescence, deregulated nutrient sensing and defects in proteostasis). Ultimately, ongoing damage and attempts at repair by continued proliferation overwhelm reparative capacity, causing loss of specialised cell functions, cell death and inflammation. This review summarises the evidence for accelerated biological ageing in atherosclerosis, the functional consequences of cell ageing on cells comprising the plaque, and the causal role that VSMC senescence plays in atherogenesis.


Subject(s)
Aging/metabolism , Atherosclerosis/metabolism , Cellular Senescence , Muscle, Smooth, Vascular/metabolism , Aging/genetics , Aging/pathology , Animals , Atherosclerosis/genetics , DNA Damage , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/growth & development , Signal Transduction
19.
Usp Fiziol Nauk ; 47(2): 45-61, 2016.
Article in Russian | MEDLINE | ID: mdl-27530043

ABSTRACT

Literature and our data suggest the regulatory action of a number of biologically active substances (catecholamines, cardiac glycosides, ß-blockers, angiotensin-converting-enzyme inhibitor) on the growth and proliferation of heart cells. By using of organotypic tissue culture has proved that the basis of this regulation is the ability of test substances, receptor- or transducer-mediated signaling to modulate the function of Na⁺, K⁺-ATPase. There is a delay in the development of vascular smooth muscle in the late postnatal period in rats with the blockade of the sympathetic nervous system in the prenatal period. The relationship between vascular remodeling and contractile activity is described. It seems that one of the causes of high blood pressure is a remodeling of the cardiovascular system, which precedes the development of hypertension.


Subject(s)
Cardiovascular System/physiopathology , Hypertension/physiopathology , Myocytes, Cardiac/metabolism , Sympathetic Nervous System/physiopathology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Cardiovascular System/growth & development , Heart/physiopathology , Humans , Hypertension/drug therapy , Hypertension/metabolism , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/pathology , Rats , Sodium-Potassium-Exchanging ATPase/metabolism
20.
Am J Physiol Heart Circ Physiol ; 309(9): H1468-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26371173

ABSTRACT

We examined the effect of stress in the first 2 wk of life induced by brief periods of daily maternal separation on developmental programming of rat small resistance mesenteric arteries (MAs). In MAs of littermate controls, mRNAs encoding mediators of vasoconstriction, including the α1a-adrenergic receptor, smooth muscle myosin heavy chain, and CPI-17, the inhibitory subunit of myosin phosphatase, increased from after birth through sexual [postnatal day (PND) 35] and full maturity, up to ∼80-fold, as measured by quantitative PCR. This was commensurate with two- to fivefold increases in maximum force production to KCl depolarization, calcium, and the α-adrenergic agonist phenylephrine, and increasing systolic blood pressure. Rats exposed to maternal separation stress as neonates had markedly accelerated trajectories of maturation of arterial contractile gene expression and function measured at PND14 or PND21 (weaning), 1 wk after the end of the stress protocol. This was suppressed by the α-adrenergic receptor blocker terazosin (0.5 mg·kg ip(-1)·day(-1)), indicating dependence on stress activation of sympathetic signaling. Due to the continued maturation of MAs in control rats, by sexual maturity (PND35) and into adulthood, no differences were observed in arterial function or response to a second stressor in rats stressed as neonates. Thus early life stress misprograms resistance artery smooth muscle, increasing vasoconstrictor function and blood pressure. This effect wanes in later stages, suggesting plasticity during arterial maturation. Further studies are indicated to determine whether stress in different periods of arterial maturation may cause misprogramming persisting through maturity and the potential salutary effect of α-adrenergic blockade in suppression of this response.


Subject(s)
Blood Pressure/genetics , Gene Expression Regulation, Developmental , Maternal Deprivation , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism , Stress, Psychological/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Blood Pressure/drug effects , Mesenteric Arteries/growth & development , Muscle Proteins/genetics , Muscle, Smooth, Vascular/growth & development , Myosin Heavy Chains/genetics , Myosin-Light-Chain Kinase/genetics , Phenylephrine/pharmacology , Phosphoproteins/genetics , Prazosin/analogs & derivatives , Prazosin/pharmacology , Protein Phosphatase 1/genetics , Rats , Receptors, Adrenergic, alpha-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Psychological/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL