Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 381
Filter
1.
Int Angiol ; 43(2): 229-239, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38619205

ABSTRACT

BACKGROUND: Varicose veins affect approximately 25% of people in industrialized countries. METHODS: The study aimed at detecting apoptotic cells and histopathological changes in varicose vein walls. Patients (N.=41) with varicose veins and 30 control group patients were divided into two groups according to their age (younger and older than 50 years). Apoptosis was determined by the TUNEL assay, elastin and collagen IV expression by immunohistochemistry and ultrastructural changes by transmission electron microscopy. RESULTS: The results show that the number of apoptotic cells in the layers of varicose veins increased, in particular in a group of patients aged over 50 years. In the varicose veins as compared to control veins the elastic fibers were found to be thinner, more fragmented and disorderly arranged. Elastin and collagen IV expression was found to decline in the intima and the media of varicose veins in both age groups. Electron microscopy demonstrated hypertrophy and degeneration of smooth muscle cells. Furthermore, cells with ultrastructural feature of apoptosis were noted. In the disorganized and expanded extracellular matrix membrane-bound vesicles, ghost bodies with different size and electron density were observed. Ghost bodies seem to bud off from smooth muscle cells and are likely to be involved in extracellular matrix remodeling as they are seen in close contact with collagen fibers. CONCLUSIONS: The study demonstrates increase of apoptotic cells in the wall of varicose veins along with vein wall structural abnormalities including alterations of smooth muscle cells and decline of elastin and collagen IV expression.


Subject(s)
Apoptosis , Elastin , Microscopy, Electron, Transmission , Myocytes, Smooth Muscle , Saphenous Vein , Varicose Veins , Humans , Saphenous Vein/ultrastructure , Saphenous Vein/pathology , Saphenous Vein/metabolism , Middle Aged , Elastin/metabolism , Varicose Veins/pathology , Varicose Veins/metabolism , Female , Adult , Male , Myocytes, Smooth Muscle/ultrastructure , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Aged , Case-Control Studies , Collagen Type IV/metabolism , Muscle, Smooth, Vascular/ultrastructure , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Immunohistochemistry , Venous Insufficiency/pathology , Venous Insufficiency/metabolism , Young Adult , Age Factors , Elastic Tissue/ultrastructure , Elastic Tissue/metabolism , Elastic Tissue/pathology
2.
Acta Neuropathol ; 142(3): 449-474, 2021 09.
Article in English | MEDLINE | ID: mdl-34309761

ABSTRACT

Parkinson's disease (PD), Parkinson's disease with dementia (PDD) and dementia with Lewy bodies (DLB) are three clinically, genetically and neuropathologically overlapping neurodegenerative diseases collectively known as the Lewy body diseases (LBDs). A variety of molecular mechanisms have been implicated in PD pathogenesis, but the mechanisms underlying PDD and DLB remain largely unknown, a knowledge gap that presents an impediment to the discovery of disease-modifying therapies. Transcriptomic profiling can contribute to addressing this gap, but remains limited in the LBDs. Here, we applied paired bulk-tissue and single-nucleus RNA-sequencing to anterior cingulate cortex samples derived from 28 individuals, including healthy controls, PD, PDD and DLB cases (n = 7 per group), to transcriptomically profile the LBDs. Using this approach, we (i) found transcriptional alterations in multiple cell types across the LBDs; (ii) discovered evidence for widespread dysregulation of RNA splicing, particularly in PDD and DLB; (iii) identified potential splicing factors, with links to other dementia-related neurodegenerative diseases, coordinating this dysregulation; and (iv) identified transcriptomic commonalities and distinctions between the LBDs that inform understanding of the relationships between these three clinical disorders. Together, these findings have important implications for the design of RNA-targeted therapies for these diseases and highlight a potential molecular "window" of therapeutic opportunity between the initial onset of PD and subsequent development of Lewy body dementia.


Subject(s)
Gene Expression Profiling/methods , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Pathology, Molecular/methods , Aged , Alternative Splicing , Alzheimer Disease , Biological Specimen Banks , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Gyrus Cinguli/pathology , Humans , Lewy Bodies/pathology , Microglia/pathology , Microglia/ultrastructure , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/ultrastructure , Parkinson Disease , RNA/genetics , Transcriptome
3.
Sci Rep ; 11(1): 13185, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162971

ABSTRACT

Medial degeneration is a common histopathological finding in aortopathy and is considered a mechanism for dilatation. We investigated if medial degeneration is specific for sporadic thoracic aortic aneurysms versus nondilated aortas. Specimens were graded by pathologists, blinded to the clinical diagnosis, according to consensus histopathological criteria. The extent of medial degeneration by qualitative (semi-quantitative) assessment was not specific for aneurysmal compared to nondilated aortas. In contrast, blinded quantitative assessment of elastin amount and medial cell number distinguished aortic aneurysms and referent specimens, albeit with marked overlap in results. Specifically, the medial fraction of elastin decreased from dilution rather than loss of protein as cross-sectional amount was maintained while the cross-sectional number, though not density, of smooth muscle cells increased in proportion to expansion of the media. Furthermore, elastic lamellae did not thin and interlamellar distance did not diminish as expected for lumen dilatation, implying a net gain of lamellar elastin and intralamellar cells or extracellular matrix during aneurysmal wall remodeling. These findings support the concepts that: (1) medial degeneration need not induce aortic aneurysms, (2) adaptive responses to altered mechanical stresses increase medial tissue, and (3) greater turnover, not loss, of mural cells and extracellular matrix associates with aortic dilatation.


Subject(s)
Aorta/anatomy & histology , Aortic Aneurysm, Thoracic/pathology , Tunica Media/ultrastructure , Adaptation, Physiological , Adult , Aged , Aorta/chemistry , Bicuspid Aortic Valve Disease/pathology , Cell Count , Comorbidity , Elastin/analysis , Extracellular Matrix/ultrastructure , Female , Humans , Male , Middle Aged , Myocytes, Smooth Muscle/ultrastructure , Single-Blind Method , Staining and Labeling , Vascular Remodeling
4.
PLoS One ; 16(5): e0246221, 2021.
Article in English | MEDLINE | ID: mdl-33999919

ABSTRACT

Due to an increasing number of cardiovascular diseases, artificial heart valves and blood vessels have been developed. Although cardiovascular applications using decellularized tissue have been studied, the mechanisms of their functionality remain unknown. To determine the important factors for preparing decellularized cardiovascular prostheses that show good in vivo performance, the effects of the luminal surface structure of the decellularized aorta on thrombus formation and cell behavior were investigated. Various luminal surface structures of a decellularized aorta were prepared by heating, drying, and peeling. The luminal surface structure and collagen denaturation were evaluated by immunohistological staining, collagen hybridizing peptide (CHP) staining, and scanning electron microscopy (SEM) analysis. To evaluate the effects of luminal surface structure of decellularized aorta on thrombus formation and cell behavior, blood clotting tests and recellularization of endothelial cells and smooth muscle cells were performed. The results of the blood clotting test showed that the closer the luminal surface structure is to the native aorta, the higher the anti-coagulant property. The results of the cell seeding test suggest that vascular cells recognize the luminal surface structure and regulate adhesion, proliferation, and functional expression accordingly. These results provide important factors for preparing decellularized cardiovascular prostheses and will lead to future developments in decellularized cardiovascular applications.


Subject(s)
Aorta/ultrastructure , Cardiovascular Diseases/diagnostic imaging , Collagen/ultrastructure , Extracellular Matrix/ultrastructure , Tissue Engineering , Animals , Aorta/pathology , Blood Vessels/pathology , Blood Vessels/ultrastructure , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Collagen/chemistry , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Extracellular Matrix/genetics , Heart Valve Prosthesis , Humans , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/ultrastructure , Swine , Thrombosis/pathology , Tissue Scaffolds
5.
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
6.
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
7.
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
8.
Reprod Fertil Dev ; 33(4): 279-290, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33573715

ABSTRACT

The myometrium goes through physiological, cellular and molecular alterations during gestation that necessitate effective cellular proteostasis. Inducible heat shock protein A1A (HSPA1A) is a member of the 70-kDa heat shock protein A (HSPA) family, which acts as a chaperone to regulate proteostasis; however, HSPA1A also participates as a cytokine in inflammatory regulation, leading to its designation as a chaperokine. This study examined the spatiotemporal expression of HSPA1A protein in the rat myometrium throughout gestation and assessed whether it is secreted as cargo of myometrial cell-derived extracellular vesicles (EVs). Immunoblot analysis demonstrated that HSPA1A expression was markedly elevated during late pregnancy and labour and increased by uterine distension. Myometrial HSPA1A expression insitu increased in myocytes of longitudinal and circular muscle layers from Day 19 through to postpartum, specifically in the cytoplasm and nuclei of myocytes from both muscle layers, but frequently detectable just outside myocyte membranes. Scanning electron microscopy examination of samples isolated from hTERT-HM cell-conditioned culture medium, using EV isolation spin columns, confirmed the presence of EVs. EV lysates contained HSPA8, HSPA1A and the EV markers apoptosis-linked gene 2-interacting protein X (Alix), the tetraspanin cluster of differentiation 63 (CD63), tumour susceptibility gene 101 (TSG101) and HSP90, but not the endoplasmic reticulum protein calnexin. These results indicate that HSPA1A may act as a chaperokine in the myometrium during pregnancy.


Subject(s)
Extracellular Vesicles/metabolism , HSP70 Heat-Shock Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Uterine Contraction , Animals , Cell Line , Extracellular Vesicles/ultrastructure , Female , Gestational Age , HSP70 Heat-Shock Proteins/genetics , Humans , Myocytes, Smooth Muscle/ultrastructure , Myometrium/ultrastructure , Pregnancy , Proteostasis , Rats, Sprague-Dawley , Up-Regulation
9.
Neurogastroenterol Motil ; 33(3): e13993, 2021 03.
Article in English | MEDLINE | ID: mdl-33020982

ABSTRACT

BACKGROUND AND AIM: Muscularis macrophages (MMs) not only mediate the innate immunity, but also functionally interact with cells important for gastrointestinal motility. The aim of this study was to determine the spatial relationship and types of contacts between the MMs and neighboring cells in the muscularis propria of human and mouse stomach, small intestine, and large intestine. METHODS: The distribution and morphology of MMs and their contacts with other cells were investigated by immunohistochemistry and transmission electron microscopy. KEY RESULTS: Immunohistochemistry showed variable shape and number of MMs according to their location in different portions of the muscle coat. By double labeling, a close association between MMs and neighboring cells, that is, neurons, smooth muscle cells, interstitial cells of Cajal (ICCs), telocytes (TCs)/PDGFRα-positive cells, was seen. Electron microscopy demonstrated that in the muscle layers of both animal species, MMs have similar ultrastructural features and have specialized cell-to-cell contacts with smooth muscle cells and TCs/PDGFRα-positive cells but not with ICCs and enteric neurons. CONCLUSION & INFERENCES: This study describes varying patterns of distribution of MMs between different regions of the gut, and reports the presence of distinct and extended cell-to-cell contacts between MMs and smooth muscle cells and between MMs and TCs/PDGFRα-positive cells. In contrast, MMs, although close to ICCs and nerve elements, did not make contact with them. These findings indicate specialized and variable roles for MMs in the modulation of gastrointestinal motility whose significance should be more closely investigated in normal and pathological conditions.


Subject(s)
Gastric Mucosa/cytology , Intercellular Junctions/ultrastructure , Intestinal Mucosa/cytology , Macrophages/cytology , Myocytes, Smooth Muscle/cytology , Telocytes/cytology , Animals , Cell Communication , Enteric Nervous System , Female , Gastric Mucosa/metabolism , Gastric Mucosa/ultrastructure , Humans , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/metabolism , Interstitial Cells of Cajal/ultrastructure , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Macrophages/metabolism , Macrophages/ultrastructure , Male , Mice , Microscopy, Electron, Transmission , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Telocytes/metabolism , Telocytes/ultrastructure
10.
Autophagy ; 17(4): 980-1000, 2021 04.
Article in English | MEDLINE | ID: mdl-32160082

ABSTRACT

Vascular smooth muscle cells (VSMCs) are an important source of foam cells in atherosclerosis. The mechanism for VSMC-derived foam cell formation is, however, poorly understood. Here, we demonstrate that the P2RY12/P2Y12 receptor is important in regulating macroautophagy/autophagy and VSMC-derived foam cell formation in advanced atherosclerosis. Inhibition of the P2RY12 receptor ameliorated lipid accumulation and VSMC-derived foam cell formation in high-fat diet-fed apoe-/- mice (atherosclerosis model) independent of LDL-c levels. Activation of the P2RY12 receptor blocked cholesterol efflux via PI3K-AKT, while genetic knockdown or pharmacological inhibition of the P2RY12 receptor inhibited this effect in VSMCs. Phosphoproteomic analysis showed that the P2RY12 receptor regulated the autophagy pathway in VSMCs. Additionally, activation of the P2RY12 receptor inhibited MAP1LC3/LC3 maturation, SQSTM1 degradation, and autophagosome formation in VSMCs. Genetic knockdown of the essential autophagy gene Atg5 significantly attenuated P2RY12 receptor inhibitor-induced cholesterol efflux in VSMCs. Furthermore, activation of the P2RY12 receptor led to the activation of MTOR through PI3K-AKT in VSMCs, whereas blocking MTOR activity (rapamycin) or reducing MTOR expression reversed the inhibition of cholesterol efflux mediated by the P2RY12 receptor in VSMCs. In vivo, inhibition of the P2RY12 receptor promoted autophagy of VSMCs through PI3K-AKT-MTOR in advanced atherosclerosis in apoe-/- mice, which could be impeded by an autophagy inhibitor (chloroquine). Therefore, we conclude that activation of the P2RY12 receptor decreases cholesterol efflux and promotes VSMC-derived foam cell formation by blocking autophagy in advanced atherosclerosis. Our study thus suggests that the P2RY12 receptor is a therapeutic target for treating atherosclerosis.Abbreviations: 2-MeSAMP: 2-methylthioadenosine 5'-monophosphate; 8-CPT-cAMP: 8-(4-chlorophenylthio)-adenosine-3',5'-cyclic-monophosphate; ABCA1: ATP binding cassette subfamily A member 1; ABCG1: ATP binding cassette subfamily G member 1; ACTB: actin beta; ADPßs: adenosine 5'-(alpha, beta-methylene) diphosphate; ALs: autolysosomes; AMPK: AMP-activated protein kinase; APOA1: apolipoprotein A1; APs: autophagosomes; ATG5: autophagy related 5; ATV: atorvastatin; AVs: autophagic vacuoles; CD: chow diet; CDL: clopidogrel; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; dbcAMP: dibutyryl-cAMP; DIL-oxLDL: dioctadecyl-3,3,3,3-tetramethylin docarbocyanine-oxLDL; EIF4EBP1/4E-BP1: eukaryotic translation initiation factor 4E binding protein 1; EVG: elastic van gieson; HE: hematoxylin-eosin; HDL: high-density lipoprotein; HFD: high-fat diet; KEGG: Kyoto Encyclopedia of Genes and Genomes; LDL-c: low-density lipoprotein cholesterol; LDs: lipid droplets; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; Masson: masson trichrome; MCPT: maximal carotid plaque thickness; MK2206: MK-2206 2HCL; NBD-cholesterol: 22-(N-[7-nitrobenz-2-oxa-1,3-diazol-4-yl] amino)-23,24-bisnor-5-cholen-3ß-ol; OLR1/LOX-1: oxidized low density lipoprotein receptor 1; ORO: oil Red O; ox-LDL: oxidized low-density lipoprotein; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TIC: ticagrelor; ULK1: unc-51 like autophagy activating kinase 1; VSMCs: vascular smooth muscle cells.


Subject(s)
Atherosclerosis/pathology , Autophagy , Foam Cells/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Receptors, Purinergic P2Y12/metabolism , Animals , Atorvastatin/pharmacology , Autophagy/drug effects , Cholesterol/metabolism , Clopidogrel/pharmacology , Drug Synergism , Female , Foam Cells/drug effects , Foam Cells/metabolism , Foam Cells/ultrastructure , Humans , Lipolysis/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
11.
Nat Protoc ; 16(1): 472-496, 2021 01.
Article in English | MEDLINE | ID: mdl-33299155

ABSTRACT

Mural cells (smooth muscle cells and pericytes) are integral components of brain blood vessels that play important roles in vascular formation, blood-brain barrier maintenance, and regulation of regional cerebral blood flow (rCBF). These cells are implicated in conditions ranging from developmental vascular disorders to age-related neurodegenerative diseases. Here we present complementary tools for cell labeling with transgenic mice and organic dyes that allow high-resolution intravital imaging of the different mural cell subtypes. We also provide detailed methodologies for imaging of spontaneous and neural activity-evoked calcium transients in mural cells. In addition, we describe strategies for single- and two-photon optogenetics that allow manipulation of the activity of individual and small clusters of mural cells. Together with measurements of diameter and flow in individual brain microvessels, calcium imaging and optogenetics allow the investigation of pericyte and smooth muscle cell physiology and their role in regulating rCBF. We also demonstrate the utility of these tools to investigate mural cells in the context of Alzheimer's disease and cerebral ischemia mouse models. Thus, these methods can be used to reveal the functional and structural heterogeneity of mural cells in vivo, and allow detailed cellular studies of the normal function and pathophysiology of mural cells in a variety of disease models. The implementation of this protocol can take from several hours to days depending on the intended applications.


Subject(s)
Brain/blood supply , Myocytes, Smooth Muscle/cytology , Optogenetics/methods , Pericytes/cytology , Animals , Blood Circulation , Female , Male , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Optical Imaging/methods , Pericytes/metabolism , Pericytes/ultrastructure
12.
Microsc Microanal ; 27(1): 227-235, 2021 02.
Article in English | MEDLINE | ID: mdl-33353579

ABSTRACT

Interstitial cells of Cajal (ICC) play an essential role in the motility of the gastrointestinal tract, and they have been identified in many laboratory animals and in humans. However, the information of ICC in lower animals is still very limited. In the present study, ICC were identified in the gastric muscularis mucosae of an amphibian­the Chinese giant salamander, by c-Kit immunohistochemistry and transmission electron microscopy. ICC showed c-Kit immunoreactivity and had spindle-shaped cell bodies and 1­2 long processes. ICC were located between smooth muscle cells (SMC) in gastric muscularis mucosae. Ultrastructurally, ICC appeared as polygon-, spindle-, and awl-shaped with long cytoplasmic prolongations between SMC. ICC had distinctive characteristics, such as nuclei with peripheral electron-dense heterochromatin, caveolae, and abundant intracytoplasmatic vacuoles, mitochondria, and rough endoplasmic reticula. Moreover, lamellar bodies and two types of condensed granules were observed in the cytoplasm of ICC. Notably, ICC establish close contacts with each other. Moreover, ICC establish gap junctions with SMC. In addition, ICC were frequently observed close to nerve fibers. In summary, the present study demonstrated the presence of ICC in the gastric muscularis mucosae of the Chinese giant salamander.


Subject(s)
Interstitial Cells of Cajal , Myocytes, Smooth Muscle , Nerve Fibers , Animals , China , Interstitial Cells of Cajal/ultrastructure , Mucous Membrane/cytology , Myocytes, Smooth Muscle/ultrastructure , Nerve Fibers/ultrastructure , Urodela
13.
Life Sci ; 264: 118714, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33157088

ABSTRACT

AIMS: The cross talk between autophagy and apoptosis of vascular smooth muscle cells (VSMCs) plays a vital role in the development of atherosclerosis (AS). Paeonol is isolated from the radix of Cortex Moutan with anti-atherosclerotic and anti-apoptosis effects. However, the mechanisms of paeonol on VSMCs apoptosis are still not fully understood. In this study, we aimed to explore whether paeonol could inhibit VSMCs apoptosis though modulating VSMCs autophagy. MATERIALS AND METHODS: The proteins expressions were detected by western blotting. Autophagosomes and apoptoticbody formation in VSMCs was observed by transmission electron microscopy (TEM). VSMCs autophagy was detected by monodansylcadaverine (MDC) staining using fluorescence microscopy, while VSMCs apoptosis was determined by 4',6-diamidino-2-phenylindole (DAPI) and flow cytometry. KEY FINDINGS: We found that paeonol could significantly increase LC3II protein level, decrease p62 and cleaved caspase-3 proteins levels in aorta of AS mice and ox-LDL-injured VSMCs. Paeonol could augment the number of autophagosomes and reduce the amount of apoptotic bodies in ox-LDL-injured VSMCs. Moreover, paeonol obviously induced VSMCs autophagy compared to ox-LDL group and remarkably suppressed VSMCs apoptosis. However, the effects of paeonol on VSMCs apoptosis could be reversed obviously by 3-MA, the autophagy inhibitor. Furthermore, paeonol could activate class III PI3K-Beclin-1 pathway significantly. Gene silencing of either class III PI3K or Beclin-1 could reverse the effects of paeonol on VSMCs autophagy and apoptosis. SIGNIFICANCE: Based on our results, paeonol could induce VSMCs autophagy by activating class III PI3K/Beclin-1 signaling pathway, thus ultimately inhibiting VSMCs apoptosis.


Subject(s)
Acetophenones/pharmacology , Apoptosis/drug effects , Autophagy , Beclin-1/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Phosphatidylinositol 3-Kinases/metabolism , Up-Regulation , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Autophagy/drug effects , Caspase 3/metabolism , Lipoproteins, LDL , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
14.
Sci Rep ; 10(1): 21045, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273629

ABSTRACT

Mitochondria are dynamic organelles that change morphology to adapt to cellular energetic demands under both physiological and stress conditions. Cardiomyopathies and neuronal disorders are associated with structure-related dysfunction in mitochondria, but three-dimensional characterizations of the organelles are still lacking. In this study, we combined high-resolution imaging and 3D electron density information provided by cryo-soft X-ray tomography to characterize mitochondria cristae morphology isolated from murine. Using the linear attenuation coefficient, the mitochondria were identified (0.247 ± 0.04 µm-1) presenting average dimensions of 0.90 ± 0.20 µm in length and 0.63 ± 0.12 µm in width. The internal mitochondria structure was successfully identified by reaching up the limit of spatial resolution of 35 nm. The internal mitochondrial membranes invagination (cristae) complexity was calculated by the mitochondrial complexity index (MCI) providing quantitative and morphological information of mitochondria larger than 0.90 mm in length. The segmentation to visualize the cristae invaginations into the mitochondrial matrix was possible in mitochondria with MCI ≥ 7. Altogether, we demonstrated that the MCI is a valuable quantitative morphological parameter to evaluate cristae modelling and can be applied to compare healthy and disease state associated to mitochondria morphology.


Subject(s)
Imaging, Three-Dimensional/methods , Mitochondria, Muscle/ultrastructure , X-Ray Microtomography/methods , Animals , Cells, Cultured , Cryopreservation/methods , Imaging, Three-Dimensional/standards , Limit of Detection , Myocytes, Smooth Muscle/ultrastructure , Rats , X-Ray Microtomography/standards
15.
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
16.
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
17.
J Cell Mol Med ; 24(12): 6658-6669, 2020 06.
Article in English | MEDLINE | ID: mdl-32337837

ABSTRACT

miR-145, the most abundant miRNA in the vascular smooth muscle cells (VSMCs), regulates VSMC function in intimal hyperplasia. It has been reported that autophagy participates in the regulation of proliferation and migration of VSMCs. However, the effect of miR-145 on autophagy and related mechanism in the proliferation and migration of VSMCs remains unclear. Therefore, we aimed to determine the effect of miR-145 on autophagy and the mechanism in VSMCs. Cell autophagy was determined by transmission electron microscope, mRFP-GFP-LC3 assay and Western blotting. A recombinant lentivirus containing miR-145 was used to construct VSMCs with miR-145 overexpression. We found that miR-145 expression was decreased, and autophagy was increased in the carotid arteries of C57BL/6J mice with intimal hyperplasia and TGF-ß1-stimulated VSMCs. Furthermore, miR-145 overexpression inhibited cell autophagy, whereas miR-145 inhibition promoted autophagy in TGF-ß1-stimulated VSMCs. Meanwhile, miR-145 inhibited the proliferation and migration of VSMCs. More importantly, our study showed that autophagy inhibition augmented the inhibitory effect of miR-145 on the proliferation and migration of VSMCs. In addition, we found that the sirtuins are not direct targets of miR-145 in the proliferation and migration of VSMCs. These results suggest that miR-145 inhibits the proliferation and migration of VSMCs by suppressing the activation of autophagy.


Subject(s)
Autophagy , Cell Movement , MicroRNAs/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Carotid Arteries/pathology , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Hyperplasia , Luciferases/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/ultrastructure , Rats, Sprague-Dawley , Sirtuins/metabolism , Transforming Growth Factor beta1/pharmacology , Tunica Intima/pathology
18.
Am J Physiol Endocrinol Metab ; 318(6): E930-E942, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32343611

ABSTRACT

Preterm birth is one of the most common complications during human pregnancy and is associated with a dramatic switch within the uterus from quiescence to contractility. However, the mechanisms underlying uterine remodeling are largely unknown. Protein kinases and phosphatases play critical roles in regulating the phosphorylation of proteins involved in the smooth muscle cell functions. In the present study, we found that Src-homology phosphatase type-1 (SHP-1, PTPN6) was significantly decreased in human myometrium in labor compared with that not in labor. Timed-pregnant mice injected intraperitoneally with the specific SHP-1 inhibitor protein tyrosine phosphatase inhibitor I (PTPI-1) manifested significantly preterm labor, with enriched plasmalemmal dense plaques between myometrial cells and increased phosphorylation at Tyr397 and Tyr576/577 sites of focal adhesion kinase (FAK) in myometrial cells, which remained to the time of labor, whereas the phosphorylation levels of ERK1/2 and phosphatidylinositol 3 kinase (PI3K) showed a rapid increase upon PTPI-1 injection but fell back to normal at the time of labor. The Tyr576/577 in FAK played an important role in the interaction between FAK and SHP-1. Knockdown of SHP-1 dramatically increased the spontaneous contraction of human uterine smooth muscle cells (HUSMCs), which was reversed by coinfection of a FAK-knockdown lentivirus. PGF2α downregulated SHP-1 via PLCß-PKC-NF-κB or PI3K-NF-κB pathways, suggesting the regenerative downregulation of SHP-1 enhances the uterine remodeling and plasticity by activating FAK and subsequent focal adhesion pathway, which eventually facilitates myometrium contraction and leads to labor. The study sheds new light on understanding of mechanisms that underlie the initiation of labor, and interventions for modulation of SHP-1 may provide a potential strategy for preventing preterm birth.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Labor, Obstetric/metabolism , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Adult , Animals , Dinoprost/pharmacology , Enzyme Inhibitors/pharmacology , Female , Focal Adhesions/ultrastructure , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/ultrastructure , Myometrium/cytology , Myometrium/drug effects , Myometrium/ultrastructure , NF-kappa B/metabolism , Obstetric Labor, Premature , Oxytocics/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C beta/metabolism , Pregnancy , Protein Kinase C/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors
19.
Ultrastruct Pathol ; 44(1): 2-14, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-32154752

ABSTRACT

It wasn't until 1960 that the dense bodies of the peripheral actin arrays of fibroblasts were finally visualized, i.e., stress fibers (SFs). Mistakenly assumed that its SFs turned the fibroblast into a unique cell situated somewhere in a continuum between it and a smooth muscle cell (SMC), it was descriptively named a "myofibroblast" (MF). Automatically, spindle cells with SFs and/or smooth muscle actin by SMA IHC-staining, became MFs, although endothelial cells, pericytes, modified SMCs (mSMC), and myoepithelial cells all contain SFs. An invisible "intermediate" cell was hypothesized to exist somewhere between SMA-negative and positive fibroblasts, and named a "proto-myofibroblast". The sub-epithelial spindle cells of normal and malignant tumors of the GI, GU, and respiratory tracts are all fibroblasts with SFs. The second erroneous myofibroblast came from a 1971 rat wound healing study and its 1974 human counterpart. Updated analysis of the papers' TEMs proved that the cells are mSMCs and not fibroblasts (AKA: MFs). The pathognomonic cells of Dupuytren's contracture are mSMCs and fibroblasts and that of the stenosing arteriopathy of Kawasaki Disease and other similar arteriopathies are mSMCs. TEM remains a powerful tool.


Subject(s)
Fibroblasts/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Animals , Arteries/pathology , Arteries/ultrastructure , Carcinoma/pathology , Dupuytren Contracture/pathology , Humans , Microscopy, Electron, Transmission , Mucocutaneous Lymph Node Syndrome/pathology , Pathologists , Tumor Microenvironment , Wound Healing/physiology
20.
Sci Rep ; 10(1): 2685, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060298

ABSTRACT

The aim of our study was to develop a novel approach to investigating mouse detrusor smooth muscle cell (SMC) physiological activity, utilizing an acute tissue dissection technique and confocal calcium imaging. The bladder of a sacrificed adult female NMRI mouse was dissected. We used light and transmission electron microscopy to assess morphology of SMCs within the tissue. Calcium imaging in individual SMCs was performed using confocal microscopy during stimulation with increasing concentrations of carbamylcholine (CCh). SMCs were identified according to their morphology and calcium activity. We determined several parameters describing the SMC responses: delays to response, recruitment, relative activity, and contraction of the tissue. CCh stimulation revealed three different SMC phenotypes: spontaneously active SMCs with and without CCh-enhanced activity and SMCs with CCh-induced activity only. SMCs were recruited into an active state in response to CCh-stimulation within a narrow range (1-25 µM); causing activation of virtually all SMCs. Maximum calcium activity of SMCs was at about 25 µM, which coincided with a visible tissue contraction. Finally, we observed shorter time lags before response onsets with higher CCh concentrations. In conclusion, our novel in situ approach proved to be a robust and reproducible method to study detrusor SMC morphology and physiology.


Subject(s)
Muscle Contraction/physiology , Myocytes, Smooth Muscle/ultrastructure , Urinary Bladder/ultrastructure , Animals , Calcium/metabolism , Cells, Cultured , Mice , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL