Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.531
Filter
Add more filters

Publication year range
1.
J Cell Physiol ; 236(11): 7342-7355, 2021 11.
Article in English | MEDLINE | ID: mdl-33928642

ABSTRACT

Vascular remodeling and restenosis are common complications after percutaneous coronary intervention. Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) play important roles in intimal hyperplasia-induced vascular restenosis. NK2 Homeobox 3 (Nkx2-3), a critical member of Nkx family, is involved in tissue differentiation and organ development. However, the role of Nkx2-3 in VSMCs proliferation and migration remains unknown. In this study, we used carotid balloon injury model and platelet-derived growth factor-BB (PDGF)-treated VSMCs as in vivo and in vitro experimental models. EdU assay and CCK-8 assay were used to detect cell proliferation. Migration was measured by scratch test. Hematoxylin and eosin staining and immunohistochemistry staining were used to evaluate the intimal hyperplasia. The autophagy level was detected by fluorescent mRFP-GFP-LC3 in vitro and by transmission electron microscopy in vivo. It was shown that Nkx2-3 was upregulated both in balloon injured carotid arteries and PDGF-stimulated VSMCs. Adenovirus-mediated Nkx2-3 overexpression inhibited intimal hyperplasia after balloon injury, and suppressed VSMCs proliferation and migration induced by PDGF. Conversely, silencing of Nkx2-3 by small interfering RNA exaggerated proliferation and migration of VSMCs. Furthermore, we found that Nkx2-3 enhanced autophagy level, while the autophagy inhibitor 3-MA eliminated the inhibitory effect of Nkx2-3 on VSMCs proliferation and migration both in vivo and in vitro. Moreover, Nkx2-3 promoted autophagy in VSMCs by activating the AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) signaling pathway. These results demonstrated for the first time that Nkx2-3 inhibited VSMCs proliferation and migration through AMPK/mTOR-mediated autophagy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Carotid Artery Injuries/enzymology , Cell Movement , Cell Proliferation , Homeodomain Proteins/physiology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/physiology , Animals , Autophagy/drug effects , Becaplermin/pharmacology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery Injuries/prevention & control , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Homeodomain Proteins/genetics , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/ultrastructure , Neointima , Rats, Sprague-Dawley , Signal Transduction , Transcription Factors/genetics , Vascular Remodeling
2.
Microvasc Res ; 136: 104172, 2021 07.
Article in English | MEDLINE | ID: mdl-33894273

ABSTRACT

BACKGROUND: VSMC proliferation and migration pathways play important roles in plaque formation in the vessel stenosis and re-stenosis processes. The microRNAs affect the expression of many genes that regulate these cellular processes. The aim of this study was to investigate the effects of miR-181b, miR-204, and miR-599 on the gene and protein expression levels of hematopoietic cell kinase (HCK) in VSMCs. METHODS: miR-181b, miR-204 were predicted for the suppression of HCK in the chemokine signaling pathway using bioinformatics tools. Then, the VSMCs were transfected by PEI-containing microRNAs. The HCK gene and protein expression levels were evaluated using RT-qPCR and Western blotting techniques, respectively. Moreover, the cellular proliferation and migration were evaluated by MTT and scratch assay methods. RESULTS: The miR-181b and miR-204 decreased significantly the HCK gene and (total and phosphorylated) protein expression levels. Also, the miR-599 did not show any significant effects on the HCK gene and protein levels. The data also showed that miR-181b, miR-204, and miR-599 prevent significantly the proliferation and migration of VSMCs. CONCLUSION: The downregulation of HCK by miR-181b and miR-204 suppressed the VSMC proliferation and migration.


Subject(s)
Cell Movement , Cell Proliferation , MicroRNAs/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Proto-Oncogene Proteins c-hck/metabolism , Cells, Cultured , Down-Regulation , Humans , MicroRNAs/genetics , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Proto-Oncogene Proteins c-hck/genetics , Signal Transduction
3.
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
4.
Arterioscler Thromb Vasc Biol ; 40(9): 2054-2069, 2020 09.
Article in English | MEDLINE | ID: mdl-32640907

ABSTRACT

OBJECTIVE: Increased CTSS (cathepsin S) has been reported to play a critical role in atherosclerosis progression. Both CTSS synthesis and secretion are essential for exerting its functions. However, the underlying mechanisms contributing to CTSS synthesis and secretion in atherosclerosis remain unclear. Approach and Results: In this study, we showed that nicotine activated autophagy and upregulated CTSS expression in vascular smooth muscle cells and in atherosclerotic plaques. Western blotting and immunofluorescent staining showed that nicotine inhibited the mTORC1 (mammalian target of rapamycin complex 1) activity, promoted the nuclear translocation of TFEB (transcription factor EB), and upregulated the expression of CTSS. Chromatin immunoprecipitation-qualificative polymerase chain reaction, electrophoretic mobility shift assay, and luciferase reporter assay further demonstrated that TFEB directly bound to the CTSS promoter. mTORC1 inhibition by nicotine or rapamycin promoted lysosomal exocytosis and CTSS secretion. Live cell assays and IP-MS (immunoprecipitation-mass spectrometry) identified that the interactions involving Rab10 (Rab10, member RAS oncogene family) and mTORC1 control CTSS secretion. Nicotine promoted vascular smooth muscle cell migration by upregulating CTSS, and CTSS inhibition suppressed nicotine-induced atherosclerosis in vivo. CONCLUSIONS: We concluded that nicotine mediates CTSS synthesis and secretion through regulating the autophagy-lysosomal machinery, which offers a potential therapeutic target for atherosclerosis treatment.


Subject(s)
Atherosclerosis/drug therapy , Autophagy/drug effects , Cathepsins/biosynthesis , Lysosomes/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nicotine/pharmacology , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cathepsins/genetics , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Exocytosis , Lysosomes/enzymology , Lysosomes/ultrastructure , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/ultrastructure , Secretory Pathway , Signal Transduction , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
5.
Acta Pharmacol Sin ; 42(11): 1798-1807, 2021 11.
Article in English | MEDLINE | ID: mdl-33589794

ABSTRACT

Proliferation of vascular smooth muscle cells (VSMCs) greatly contributes to vascular remodeling in hypertension. This study is to determine the roles and mechanisms of miR-135a-5p intervention in attenuating VSMC proliferation and vascular remodeling in spontaneously hypertensive rats (SHRs). MiR-135a-5p level was raised, while fibronectin type III domain-containing 5 (FNDC5) mRNA and protein expressions were reduced in VSMCs of SHRs compared with those of Wistar-Kyoto rats (WKYs). Enhanced VSMC proliferation in SHRs was inhibited by miR-135a-5p knockdown or miR-135a-5p inhibitor, but exacerbated by miR-135a-5p mimic. VSMCs of SHRs showed reduced myofilaments, increased or even damaged mitochondria, increased and dilated endoplasmic reticulum, which were attenuated by miR-135a-5p inhibitor. Dual-luciferase reporter assay shows that FNDC5 was a target gene of miR-135a-5p. Knockdown or inhibition of miR-135a-5p prevented the FNDC5 downregulation in VSMCs of SHRs, while miR-135a-5p mimic inhibited FNDC5 expressions in VSMCs of both WKYs and SHRs. FNDC5 knockdown had no significant effects on VSMC proliferation of WKYs, but aggravated VSMC proliferation of SHRs. Exogenous FNDC5 or FNDC5 overexpression attenuated VSMC proliferation of SHRs, and prevented miR-135a-5p mimic-induced enhancement of VSMC proliferation of SHR. MiR-135a-5p knockdown in SHRs attenuated hypertension, normalized FNDC5 expressions and inhibited vascular smooth muscle proliferation, and alleviated vascular remodeling. These results indicate that miR-135a-5p promotes while FNDC5 inhibits VSMC proliferation in SHRs. Silencing of miR-135a-5p attenuates VSMC proliferation and vascular remodeling in SHRs via disinhibition of FNDC5 transcription. Either inhibition of miR-135a-5p or upregulation of FNDC5 may be a therapeutically strategy in attenuating vascular remodeling and hypertension.


Subject(s)
Hypertension/metabolism , MicroRNAs/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Remodeling/physiology , Animals , Cell Proliferation/physiology , Cells, Cultured , Hypertension/pathology , Male , MicroRNAs/antagonists & inhibitors , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Rats , Rats, Inbred SHR , Rats, Inbred WKY
6.
Proc Natl Acad Sci U S A ; 115(16): E3682-E3691, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610298

ABSTRACT

Hemodynamic forces and Notch signaling are both known as key regulators of arterial remodeling and homeostasis. However, how these two factors integrate in vascular morphogenesis and homeostasis is unclear. Here, we combined experiments and modeling to evaluate the impact of the integration of mechanics and Notch signaling on vascular homeostasis. Vascular smooth muscle cells (VSMCs) were cyclically stretched on flexible membranes, as quantified via video tracking, demonstrating that the expression of Jagged1, Notch3, and target genes was down-regulated with strain. The data were incorporated in a computational framework of Notch signaling in the vascular wall, where the mechanical load was defined by the vascular geometry and blood pressure. Upon increasing wall thickness, the model predicted a switch-type behavior of the Notch signaling state with a steep transition of synthetic toward contractile VSMCs at a certain transition thickness. These thicknesses varied per investigated arterial location and were in good agreement with human anatomical data, thereby suggesting that the Notch response to hemodynamics plays an important role in the establishment of vascular homeostasis.


Subject(s)
Jagged-1 Protein/physiology , Mechanotransduction, Cellular/physiology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Receptor, Notch3/physiology , Aged , Arteries/ultrastructure , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Computer Simulation , Endothelial Cells/metabolism , Gene Expression Regulation , Homeostasis , Humans , Jagged-1 Protein/biosynthesis , Jagged-1 Protein/genetics , Ligands , Middle Aged , Models, Biological , Morphogenesis/physiology , Muscle, Smooth, Vascular/ultrastructure , Receptor, Notch3/biosynthesis , Receptor, Notch3/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Stress, Mechanical , Transcription Factor HES-1/biosynthesis , Transcription Factor HES-1/genetics , Video Recording
7.
Microvasc Res ; 128: 103938, 2020 03.
Article in English | MEDLINE | ID: mdl-31682800

ABSTRACT

OBJECTIVE: In the present study, we used a two-kidney-two-clip (2k2c) stroke-prone renovascular hypertension rat model (RHRSP) to investigate the protective effects of ligustrazine (TMP) on cerebral arteries and to examine PI3K/Akt pathway behavior under this protection. METHODS: The cerebral artery remodeling was induced by 2k2c-induced renovascular hypertension. Brain basilar artery tissues were isolated and their histological changes were detected through H&E and EVG staining, α-SMA IHC staining, and transmission electron microscopy at four, eight, and twelve weeks after 2k2c surgery, both with and without TMP treatment. Meanwhile, the ET-1, Ang II, and NO levels in basilar arteries and plasma were determined. Furthermore, the PTEN expression and the activation of PI3K/Akt in basilar artery tissues were detected through IHC and Western Blot. In addition, the primary basilar artery smooth muscle cells (BASMCs) were cultured and TMP protection of BASMCs stimulated with ET-1/Ang II in the presence or absence of insulin-like growth factor 1 (IGF-1) was determined. RESULTS: TMP attenuated basilar artery remodeling, decreased ET-1 and Ang II levels and increased NO level in basilar arteries and plasma of RHRSP rats. Moreover, TMP reduced BASMCs proliferation upon ET-1/Ang II stimulation. We also found that TMP could effectively suppress the activation of PI3K/Akt in 2k2c-RHRSP rat basilar artery and ET-1/Ang II stimulated BASMCs. Most importantly, IGF-1, as an activator of PI3K/Akt, could damage the protective effect of TMP. CONCLUSIONS: TMP exerts its protective effects and prevents basilar artery remodeling in RHRSP rats at least partly through the inhibition of PI3K/Akt pathway.


Subject(s)
Hypertension, Renovascular/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/pharmacology , Temporal Arteries/drug effects , Vascular Remodeling/drug effects , Angiotensin II/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelin-1/metabolism , Hypertension, Renovascular/enzymology , Hypertension, Renovascular/pathology , Hypertension, Renovascular/physiopathology , Ligation , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Nitric Oxide/metabolism , Rats, Sprague-Dawley , Renal Artery/surgery , Signal Transduction , Temporal Arteries/enzymology , Temporal Arteries/physiopathology , Temporal Arteries/ultrastructure
8.
Ultrastruct Pathol ; 44(1): 103-115, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906762

ABSTRACT

To clarify foam cell origination in atherosclerosis, a series of morphologic and ultrastructural alterations of vascular smooth muscle cells (VSMCs) and foam cells were studied by light and electron microscopy in atherosclerotic aortas from hyperlipidemic rabbits induced for 5 weeks. The study exhibited that VSMCs were severely degenerated and damaged, including irregular shapes, expanded mitochondria, aplenty lipid droplets, and disarranged myofilaments in cytoplasm in media adjacent to atheromatic bottoms. Most lipid laden cells shared interphase structures of VSMCs and foam cells, and some dissolved spindle cells contained lipid droplets, lipofuscin, and rod-like CCs in cytoplasm also. The result demonstrated that VSMCs were degenerated and transformed into foam cells in atherosclerosis, which was responsible for the accumulation of lipid and cholesterol crystals in atherosclerotic arteries.


Subject(s)
Atherosclerosis/pathology , Foam Cells/ultrastructure , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Animals , Aorta , Foam Cells/pathology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Rabbits
9.
Int J Mol Sci ; 21(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32423114

ABSTRACT

Vascular calcification and stiffening of the arterial wall is a systemic phenomenon that is associated with aging and it can be increased by several risk factors. The underlying mechanisms, especially the pathways of cellular senescence, are under current investigation. Easily manageable in vitro settings help to study the signaling pathways. The experimental setting presented here is based on an in vitro model using rat vascular smooth muscle cells and the detection of senescence and osteoblastic markers via immunofluorescence and RNAscope™. Co-staining of the senescence marker p21, the osteoblastic marker osteopontin, detection of senescence-associated heterochromatin foci, and senescence-associated ß-galactosidase is possible within one test approach requiring fewer cells. The protocol is a fast and reliable evaluation method for multiplexing of calcifying and senescence markers with fluorescence microscopy detection. The experimental setting enables analysis on single cell basis and allows detection of intra-individual variances of cultured cells.


Subject(s)
Osteopontin/genetics , Vascular Calcification/genetics , beta-Galactosidase/genetics , p21-Activated Kinases/genetics , Aging/genetics , Animals , Arteries/metabolism , Biomarkers/metabolism , Cellular Senescence/genetics , Humans , Microscopy, Fluorescence , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Rats , Signal Transduction/genetics , p21-Activated Kinases/metabolism
10.
J Cell Mol Med ; 23(2): 819-827, 2019 02.
Article in English | MEDLINE | ID: mdl-30450815

ABSTRACT

The blood-brain barrier (BBB) is essential for a functional neurovascular unit. Most studies focused on the cells forming the BBB, but very few studied the basement membrane (BM) of brain capillaries in ageing. We used transmission electron microscopy and electron tomography to investigate the BM of the BBB in ageing C57BL/6J mice. The thickness of the BM of the BBB from 24-month-old mice was double as compared with that of 6-month-old mice (107 nm vs 56 nm). The aged BBB showed lipid droplets gathering within the BM which further increased its thickness (up to 572 nm) and altered its structure. The lipids appeared to accumulate toward the glial side of the BM. Electron tomography showed that the lipid-rich BM regions are located in small pockets formed by the end-feet of astrocytes. These findings suggest an imbalance of the lipid metabolism and that may precede the structural alteration of the BM. These alterations may favour the accretion of abnormal proteins that lead to neurodegeneration in ageing. These findings warrant further investigation of the BM of brain capillaries and of adjoining cells as potential targets for future therapies.


Subject(s)
Aging/physiology , Basement Membrane/ultrastructure , Blood-Brain Barrier/ultrastructure , Capillaries/ultrastructure , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Basement Membrane/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/metabolism , Brain/ultrastructure , Capillaries/metabolism , Electron Microscope Tomography , Lipid Droplets/metabolism , Lipid Droplets/ultrastructure , Lipid Metabolism/physiology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neuroglia/metabolism , Neuroglia/ultrastructure
11.
J Cell Physiol ; 234(6): 8668-8682, 2019 06.
Article in English | MEDLINE | ID: mdl-30456860

ABSTRACT

Aberrant proliferation of vascular smooth muscle cells (VSMC) is a critical contributor to the pathogenesis of atherosclerosis (AS). Our previous studies have demonstrated that apelin-13/APJ confers a proliferative response in VSMC, however, its underlying mechanism remains elusive. In this study, we aimed to investigate the role of mitophagy in apelin-13-induced VSMC proliferation and atherosclerotic lesions in apolipoprotein E knockout (ApoE-/-) mice. Apelin-13 enhances human aortic VSMC proliferation and proliferative regulator proliferating cell nuclear antigen expression in dose and time-dependent manner, while is abolished by APJ antagonist F13A. We observe the engulfment of damage mitochondria by autophagosomes (mitophagy) of human aortic VSMC in apelin-13 stimulation. Mechanistically, apelin-13 increases p-AMPKα and promotes mitophagic activity such as the LC3I to LC3II ratio, the increase of Beclin-1 level and the decrease of p62 level. Importantly, the expressions of PINK1, Parkin, VDAC1, and Tom20 are induced by apelin-13. Conversely, blockade of APJ by F13A abolishes these stimulatory effects. Human aortic VSMC transfected with AMPKα, PINK1, or Parkin and subjected to apelin-13 impairs mitophagy and prevents proliferation. Additional, apelin-13 not only increases the expression of Drp1 but also reduces the expressions of Mfn1, Mfn2, and OPA1. Remarkably, the mitochondrial division inhibitor-1(Mdivi-1), the pharmacological inhibition of Drp1, attenuates human aortic VSMC proliferation. Treatment of ApoE-/- mice with apelin-13 accelerates atherosclerotic lesions, increases p-AMPKα and mitophagy in aortic wall in vivo. Finally, PINK1-/- mutant mice with apelin-13 attenuates atherosclerotic lesions along with defective in mitophagy. PINK1/Parkin-mediated mitophagy promotes apelin-13-evoked human aortic VSMC proliferation by activating p-AMPKα and exacerbates the progression of atherosclerotic lesions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Mitochondria, Muscle/drug effects , Mitophagy/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/ultrastructure , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/ultrastructure , Phosphorylation , Plaque, Atherosclerotic , Protein Kinases/deficiency , Protein Kinases/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics
12.
Am J Physiol Heart Circ Physiol ; 317(4): H867-H876, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31441677

ABSTRACT

Atherosclerosis is the most common underlying cause of cardiovascular morbidity and mortality worldwide. c-Kit (CD117) is a member of the receptor tyrosine kinase family, which regulates differentiation, proliferation, and survival of multiple cell types. Recent studies have shown that c-Kit and its ligand stem cell factor (SCF) are present in arterial endothelial cells and smooth muscle cells (SMCs). The role of c-Kit in cardiovascular disease remains unclear. The aim of the current study is to determine the role of c-Kit in atherogenesis. For this purpose, atherosclerotic plaques were quantified in c-Kit-deficient mice (KitMut) after they were fed a high-fat diet (HFD) for 16 wk. KitMut mice demonstrated substantially greater atherosclerosis compared with control (KitWT) littermates (P < 0.01). Transplantation of c-Kit-positive bone marrow cells into KitMut mice failed to rescue the atherogenic phenotype, an indication that increased atherosclerosis was associated with reduced arterial c-Kit. To investigate the mechanism, SMC organization and morphology were analyzed in the aorta by histopathology and electron microscopy. SMCs were more abundant, disorganized, and vacuolated in aortas of c-Kit mutant mice compared with controls (P < 0.05). Markers of the "contractile" SMC phenotype (calponin, SM22α) were downregulated with pharmacological and genetic c-Kit inhibition (P < 0.05). The absence of c-Kit increased lipid accumulation and significantly reduced the expression of the ATP-binding cassette transporter G1 (ABCG1) necessary for lipid efflux in SMCs. Reconstitution of c-Kit in cultured KitMut SMCs resulted in increased spindle-shaped morphology, reduced proliferation, and elevated levels of contractile markers, all indicators of their restored contractile phenotype (P < 0.05).NEW & NOTEWORTHY This study describes the novel vasculoprotective role of c-Kit against atherosclerosis and its function in the preservation of the SMC contractile phenotype.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Hyperlipidemias/complications , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-kit/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Aorta/metabolism , Aorta/ultrastructure , Aortic Diseases/etiology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hyperlipidemias/metabolism , Mice, Knockout, ApoE , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/ultrastructure , Mutation , Myocytes, Smooth Muscle/ultrastructure , Phenotype , Plaque, Atherosclerotic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction , Calponins
13.
Microcirculation ; 26(6): e12527, 2019 08.
Article in English | MEDLINE | ID: mdl-30597676

ABSTRACT

OBJECTIVE: This study was undertaken to characterize structural and pharmacological properties of the pig popliteal artery in order to develop a novel system for the examination of lower limb blood flow regulation in a variety of cardiovascular pathologies, such as diabetes-induced peripheral artery disease. METHODS: Popliteal arteries were isolated from streptozocin-induced diabetic pigs or age-matched saline-injected control pigs for morphological study using transmission electron microscopy and for examination of vasoreactivity to pharmacological agents using wire myography. RESULTS: Transmission electron microscopy of the porcine popliteal artery wall revealed the presence of endothelial cell-smooth muscle cell interactions (myoendothelial junctions) and smooth muscle cell-smooth muscle cell interactions, for which we have coined the term "myo-myo junctions." These myo-myo junctions were shown to feature plaques indicative of connexin expression. Further, the pig popliteal artery was highly responsive to a variety of vasoconstrictors including norepinephrine, phenylephrine, and U46619, and vasodilators including acetylcholine, adenosine 5'-[ß-thio] diphosphate, and bradykinin. Finally, 2 weeks after streptozocin-induced diabetes, the normalized vasoconstriction of the pig popliteal artery to norepinephrine was unaltered compared to control. CONCLUSIONS: The pig popliteal artery displays structural and pharmacological properties that might prove useful in future studies of diabetes-associated peripheral artery disease and other lower limb cardiovascular diseases.


Subject(s)
Diabetic Angiopathies , Lower Extremity/blood supply , Peripheral Arterial Disease , Popliteal Artery , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/pathology , Peripheral Arterial Disease/physiopathology , Popliteal Artery/metabolism , Popliteal Artery/physiopathology , Popliteal Artery/ultrastructure , Swine , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
14.
Cell Tissue Res ; 376(1): 137-141, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30610452

ABSTRACT

Ehlers-Danlos syndrome (EDS) is a collection of inheritable diseases involving the musculoskeletal, integumentary and visual systems. Spondylodysplastic EDS-ZIP13 (spEDS-ZIP13: OMIM 612350) was recently defined as a new form of EDS. Although vasculitis has been found in many spEDS-ZIP13 patients, vascular pathology has not been included as a pathognomonic lesion of this type of EDS. We investigate the morphometry of the thoracic aorta in wild-type and Zip13-knockout (Zip13-KO) mice. Our assessment found abnormalities in the number and morphology of elastic and cellular components in the aortic wall, especially the tunica media, of Zip13-KO mice, indicating aortic fragility. Accordingly, our major findings (vascular smooth muscle cells with small nuclei, small percentage of elastic membrane area per tunica media, many large elastic flaps) should be considered vulnerable characteristics indicating fragility of the aorta in patients with spEDS-ZIP13.


Subject(s)
Aorta, Thoracic/abnormalities , Ehlers-Danlos Syndrome/pathology , Muscle, Smooth, Vascular/abnormalities , Osteochondrodysplasias/pathology , Animals , Aorta, Thoracic/pathology , Cation Transport Proteins/genetics , Elasticity , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/ultrastructure
15.
Arterioscler Thromb Vasc Biol ; 38(10): 2423-2434, 2018 10.
Article in English | MEDLINE | ID: mdl-30354218

ABSTRACT

Objective- Actin cytoskeleton assembly and organization, as a result of focal adhesion (FA) formation during cell adhesion, are dependent on reactive oxygen species and the cellular redox environment. Poldip2 (polymerase δ-interacting protein 2), a novel regulator of NOX4 (NADPH oxidase 4), plays a significant role in reactive oxygen species production and cytoskeletal remodeling. Thus, we hypothesized that endogenous reactive oxygen species derived from Poldip2/NOX4 contribute to redox regulation of actin and cytoskeleton assembly during integrin-mediated cell adhesion. Approach and Results- Using vascular smooth muscle cells, we verified that hydrogen peroxide (H2O2) levels increase during integrin-mediated cell attachment as a result of activation of NOX4. Filamentous actin (F-actin) was oxidized by sulfenylation during cell attachment, with a peak at 3 hours (0.80±0.04 versus 0.08±0.13 arbitrary units at time zero), which was enhanced by overexpression of Poldip2. Depletion of Poldip2 or NOX4 using siRNA, or scavenging of endogenous H2O2 with catalase, inhibited F-actin oxidation by 78±26%, 99±1%, and 98±1%, respectively. To determine the consequence of F-actin oxidation, we examined the binding of F-actin to vinculin, a protein involved in FA complexes that regulates FA maturation. Vinculin binding during cell adhesion as well as migration capacity were inhibited after transfection with actin containing 2 oxidation-resistant point mutations (C272A and C374A). Silencing of Poldip2 or NOX4 also impaired actin-vinculin interaction, which disturbed maturation of FAs and inhibited cell migration. Conclusions- These results suggest that integrin engagement during cell attachment activates Poldip2/Nox4 to oxidize actin, which modulates FA assembly.


Subject(s)
Actin Cytoskeleton/enzymology , Carrier Proteins/metabolism , Cell Adhesion , Integrins/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 4/metabolism , Nuclear Proteins/metabolism , Vinculin/metabolism , Actin Cytoskeleton/genetics , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Humans , Hydrogen Peroxide/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , NADPH Oxidase 4/genetics , Nuclear Proteins/genetics , Oxidation-Reduction , Rats , Signal Transduction
16.
Arterioscler Thromb Vasc Biol ; 38(11): 2651-2664, 2018 11.
Article in English | MEDLINE | ID: mdl-30354243

ABSTRACT

Objective- Mutations affecting contractile-related proteins in the ECM (extracellular matrix), microfibrils, or vascular smooth muscle cells can predispose the aorta to aneurysms. We reported previously that the LRP1 (low-density lipoprotein receptor-related protein 1) maintains vessel wall integrity, and smLRP1-/- mice exhibited aortic dilatation. The current study focused on defining the mechanisms by which LRP1 regulates vessel wall function and integrity. Approach and Results- Isometric contraction assays demonstrated that vasoreactivity of LRP1-deficient aortic rings was significantly attenuated when stimulated with vasoconstrictors, including phenylephrine, thromboxane receptor agonist U-46619, increased potassium, and L-type Ca2+ channel ligand FPL-64176. Quantitative proteomics revealed proteins involved in actin polymerization and contraction were significantly downregulated in aortas of smLRP1-/- mice. However, studies with calyculin A indicated that although aortic muscle from smLRP1-/- mice can contract in response to calyculin A, a role for LRP1 in regulating the contractile machinery is not revealed. Furthermore, intracellular calcium imaging experiments identified defects in calcium release in response to a RyR (ryanodine receptor) agonist in smLRP1-/- aortic rings and cultured vascular smooth muscle cells. Conclusions- These results identify a critical role for LRP1 in modulating vascular smooth muscle cell contraction by regulating calcium signaling events that potentially protect against aneurysm development.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium Signaling , Cytoskeletal Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Vasoconstriction , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Animals , Aorta/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cytoskeletal Proteins/genetics , Female , Gene Expression Regulation , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Receptors, LDL/deficiency , Receptors, LDL/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Tissue Culture Techniques , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
17.
J Pathol ; 244(4): 408-420, 2018 04.
Article in English | MEDLINE | ID: mdl-29266233

ABSTRACT

Mutations in the α1 (COL4A1) or α2 (COL4A2) chains of collagen type IV, a major component of the vascular basement membrane, cause intracerebral haemorrhages with variable expressivity and reduced penetrance by mechanisms that remain poorly understood. Here we sought to investigate the cellular mechanisms of COL4A1-related intracerebral haemorrhage and identify a marker for haemorrhage risk stratification. A combination of histological, immunohistochemical, and electron microscopy analyses were used to analyse the brain parenchyma, cerebrovasculature, and retinal vessels of mice expressing the disease-causing COL4A1 p.G498V mutation. Mutant mice developed cerebral microhaemorrhages and macroscopic haemorrhages (macrohaemorrhages), the latter with reduced penetrance, mimicking the human disease. Microhaemorrhages that occurred in early postnatal life were associated with a transient, generalized increase in blood-brain barrier permeability at the level of capillaries. Macrohaemorrhages, which occurred later in life, originated from deep brain arteries with focal loss of smooth muscle cells. Similar smooth muscle cell loss was detected in retinal arteries, and a time-course analysis of arterial lesions showed that smooth muscle cells are recruited normally in arterial wall during development, but undergo progressive apoptosis-mediated degeneration. By assessing in parallel the extent of these retinal arterial lesions and the presence/absence of macrohaemorrhages, we found that the arterial lesion load in the retina is strongly correlated with the burden of macrohaemorrhages. We conclude that microhaemorrhages and macrohaemorrhages are driven by two distinct mechanisms. Moreover, smooth muscle cell degeneration is a critical factor underlying the partial penetrance of COL4A1-related macrohaemorrhages, and retinal imaging is a promising tool for identifying high-risk patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cerebral Hemorrhage/pathology , Collagen Type IV/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Stroke/pathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cell Proliferation , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Collagen Type IV/deficiency , Collagen Type IV/genetics , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Predisposition to Disease , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Penetrance , Peptide Fragments/genetics , Peptide Fragments/metabolism , Retinal Artery/metabolism , Retinal Artery/pathology , Stroke/genetics , Stroke/metabolism , Time Factors
18.
Ultrastruct Pathol ; 43(4-5): 190-198, 2019.
Article in English | MEDLINE | ID: mdl-31522593

ABSTRACT

Cardiovascular disease secondary to diabetes represents a significant challenge to the health community. The advanced glycation end products (AGEs) play an important role in diabetes-mediated vascular injury. We tested whether metformin can suppress aortic AGEs production and protect against aortic injuries (aortopathy) and hypertension in streptozotocin-induced type 2 diabetes mellitus (T2DM) animal model. T2DM was induced in rats two weeks after being fed on a high carbohydrate and fat diet (HCFD), and continued on a HCFD until being sacrificed at week 12 (model group). The protective group was put on metformin two weeks before diabetic induction and continued on metformin and HCFD until the end of the experiment, at week 12. Using electron microscopy examinations, we observed in the model group substantial damage to the ultrastructure of aortic endothelial and vascular smooth muscle layers as demonstrated by markedly distorted vacuolated endothelial and vascular smooth muscle cells with pyknotic nuclei detached from the underlying basement membrane, which were protected by metformin. Also, metformin significantly (p < .05) decreased both systolic and diastolic blood pressure, aortic levels of AGEs, and blood levels of oxidative stress and inflammatory biomarkers. We conclude that metformin protects against T2DM-induced aortopathy and hypertension, possibly via the inhibition of AGEs, inflammation, and oxidative stress.


Subject(s)
Antioxidants/pharmacology , Aorta/drug effects , Diabetes Mellitus, Type 2 , Glycation End Products, Advanced/metabolism , Metformin/pharmacology , Animals , Aorta/pathology , Aorta/ultrastructure , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Glycation End Products, Advanced/drug effects , Hypertension/etiology , Hypoglycemic Agents/pharmacology , Male , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Rats
19.
Kidney Int ; 93(2): 343-354, 2018 02.
Article in English | MEDLINE | ID: mdl-29032812

ABSTRACT

In patients with chronic kidney and end-stage renal diseases, the major risk factor for progression of arterial calcification is the presence of existing (baseline) calcification. Here, we tested whether calcification of arteries is extended from calcified vascular smooth muscle cells (VSMCs) to adjacent normal cells by matrix vesicle-induced alteration of cell signaling. Matrix vesicles isolated from VSMC of rats with chronic kidney disease were co-cultured with VSMCs from normal littermates. Endocytosis of vesicles by recipient cells was confirmed by confocal microscopy. The addition of cellular matrix vesicles with characteristics of exosomes and low fetuin-A content enhanced the calcification of recipient VSMC. Further, only cellular-derived matrix vesicles induced an increase in intracellular calcium ion concentration, NOX1 (NADPH oxidase) and the anti-oxidant superoxide dismutase-2 in recipient normal VSMC. The increase in intracellular calcium ion concentration was due to release from endoplasmic reticulum and partially attributed to the activation of both NOX1 and mitogen-activated protein kinase (MEK1 and Erk1/2) signaling, since inhibiting both pathways blocked the increase in intracellular calcium ion in recipient VSMC. In contrast, matrix vesicles isolated from the media had no effect on the intracellular calcium ion concentration or MEK1 signaling, and did not induce calcification. However, media matrix vesicles did increase Erk1/2, although not to the level of cellular matrix vesicles, and NOX1 expression. Blockade of NOX activity further inhibited the cellular matrix vesicle-induced accelerated calcification of recipient VSMC, suggesting a potential therapeutic role of such inhibition. Thus, addition of cellular-derived matrix vesicles from calcifying VSMC can accelerate calcification by inducing cell signaling changes and phenotypic alteration of recipient VSMC.


Subject(s)
Calcium/metabolism , Endocytosis , Exosomes/metabolism , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Renal Insufficiency, Chronic/metabolism , Signal Transduction , Vascular Calcification/metabolism , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Exosomes/ultrastructure , Extracellular Matrix/ultrastructure , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 1/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , NADPH Oxidase 1/metabolism , Phenotype , Rats , Renal Insufficiency, Chronic/pathology , Superoxide Dismutase/metabolism , Vascular Calcification/pathology
20.
Biochem Biophys Res Commun ; 505(1): 282-289, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30245136

ABSTRACT

Thromboangiitis obliterans (TAO), also known as Buerger's disease, is a nonatherosclerotic inflammatory disease that influences medium- and small-sized blood vessels of extremities. However, mechanisms underlying TAO are still unclear. As a mediator associated with inflammation, A disintegrin and metalloprotease 10 (ADAM10) was hypothesized to play inhibitory roles in the development of TAO. Thus, the objective of this study is to investigate the effects of ADAM10 in a sodium laurate-induced TAO rat model and elucidate underlying mechanisms. Male Wistar rats were randomly divided into four groups (n = 6) for treatment: sham-operated (SHAM), TAO model (TAO), ADAM10 low dose injection (3 mg/kg; ADAM10-LD) and ADAM10 high dose injection (6 mg/kg; ADAM10-HD). After 14-day treatment, color Doppler ultrasound and hematology analysis indicated TAO rats displayed higher whole blood viscosity and blood platelet count compared with those in the SHAM group. Histologic evaluation and transmission electron microscopy revealed that the ultrastructural damages of vascular smooth muscle and endothelial cells were observed in TAO rats, such as fractured endoplasmic reticulum, decreased cell counts, and fibrillation. On the other hand, the typical signs and symptoms of TAO rats were significantly alleviated via ADAM10 treatment with a dose-dependent pattern. Real-time PCR and western blot results revealed that the expression of high-mobility-group box 1 (HMGB1), receptor for advanced glycation end-products (RAGE) and nuclear factor-kappa B (NF-κB) increased in TAO rats whereas decreased by ADAM10 treatment in both mRNA and protein levels. In conclusion, the results suggest ADAM10 alleviates symptoms of sodium laurate-induced TAO in rats via the RAGE/NF-κB signaling pathway and provides insight into the molecular basis and a potential therapeutic strategy for TAO.


Subject(s)
ADAM10 Protein/pharmacology , HMGB1 Protein/metabolism , NF-kappa B/metabolism , Receptor for Advanced Glycation End Products/metabolism , Thromboangiitis Obliterans/prevention & control , ADAM10 Protein/administration & dosage , Animals , Blood Viscosity/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , HMGB1 Protein/genetics , Lauric Acids , Male , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/ultrastructure , NF-kappa B/genetics , Platelet Count , Rats, Wistar , Receptor for Advanced Glycation End Products/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thromboangiitis Obliterans/blood , Thromboangiitis Obliterans/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL