Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 963
Filter
1.
Phytochemistry ; 211: 113700, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37119920

ABSTRACT

Plumula Nelumbinis, the embryo of the seed of Nelumbo nucifera Gaertn, is commonly used to make tea and nutritional supplements in East Asian countries. A bioassay-guided isolation of Plumula Nelumbinis afforded six previously undescribed bisbenzylisoquinoline alkaloids, as well as seven known alkaloids. Their structures were elucidated by extensive analysis of HRESIMS, NMR, and CD data. Pycnarrhine, neferine-2α,2'ß-N,N-dioxides, neferine, linsinine, isolinsinine, and nelumboferine, at 2 µM significantly suppressed the migration of MOVAS cells with inhibition ratio above 50%, more active than that of the positive control cinnamaldehyde (inhibition ratio 26.9 ± 4.92%). Additionally, neferine, linsinine, isolinsinine, and nelumboferine, were also active against the proliferation of MOVAS cells with inhibition ratio greater than 45%. The preliminary structure-activity relationships were discussed. Mechanism studies revealed that nelumboferine inhibited the migration and proliferation of MOVAS cells by regulating ORAI2/Akt signaling pathway.


Subject(s)
Alkaloids , Benzylisoquinolines , Proto-Oncogene Proteins c-akt , Muscle, Smooth, Vascular/chemistry , Alkaloids/chemistry , Benzylisoquinolines/pharmacology , Cell Proliferation
2.
Clin Exp Nephrol ; 26(12): 1160-1169, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36070106

ABSTRACT

BACKGROUND: The accumulation of glucose degradation products (GDPs) during peritoneal dialysis (PD) can lead to immature angiogenesis in the peritoneum. However, the effect of GDPs on angiogenesis, at concentrations observed in dialysate effluent, has not been widely investigated. We do not know how the inflammation observed in PD-related peritonitis affects angiogenesis of the peritoneum. METHODS: Human umbilical vessel endothelial cells (HUVEC) and human umbilical aortic smooth muscle cells (HUASMC) were used to examine the response to the three main GDPs found in peritoneal dialysate (methylglyoxal (MGO), 3-deoxyglucosone (3-DG), and 5-hydroxymethylfurfural (5-HMF). Supernatant from lipopolysaccharide (LPS)-activated murine macrophage cell lines (RAW 264.7 cells) were used to stimulate angiogenesis in the peritoneum. Changes in the expression of vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor B (PDGFB) in HUVEC, and PDGF-receptor beta (PDGF-Rß) in HUASMC, were examined by real-time PCR, Western blot, and ELISA. RESULTS: In HUVECs, the expression of PDGFB mRNA and protein were decreased by exposure to MGO, 3-DG, and 5-HMF at concentrations observed in dialysate effluent. A subsequent decrease in secreted PDGF-BB was observed. In HUASMCs, MGO and 5-HMF increased the expression of VEGF-A mRNA and protein, while 5-HMF decreased the expression of PDGF-Rß. VEGF-A is upregulated, and PDGF-Rß is downregulated, by conditioned medium of LPS-stimulated macrophages in HUASMCs. CONCLUSIONS: The GDPs of PD effluent cause an imbalance of angiogenic factors in endothelial cells and vascular smooth muscle cells that may lead to immature angiogenesis in the peritoneum.


Subject(s)
Angiogenesis Inducing Agents , Dialysis Solutions , Glucose , Neovascularization, Pathologic , Peritoneal Dialysis , Animals , Humans , Mice , Dialysis Solutions/pharmacology , Endothelial Cells/metabolism , Glucose/pharmacology , Glucose/metabolism , Lipopolysaccharides/pharmacology , Magnesium Oxide/metabolism , Muscle, Smooth, Vascular/chemistry , Myocytes, Smooth Muscle/chemistry , Neovascularization, Pathologic/metabolism , Peritoneal Dialysis/adverse effects , Peritoneum/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Pyruvaldehyde/pharmacology , Pyruvaldehyde/metabolism , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism , Human Umbilical Vein Endothelial Cells , RAW 264.7 Cells
3.
Part Fibre Toxicol ; 19(1): 29, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35449013

ABSTRACT

BACKGROUND: Epidemiological studies have suggested that elevated concentrations of particulate matter (PM) are strongly associated with the incidence of atherosclerosis, however, the underlying cellular and molecular mechanisms of atherosclerosis by PM exposure and the components that are mainly responsible for this adverse effect remain to be established. In this investigation, we evaluated the effects of ambient PM on vascular smooth muscle cell (VSMC) behavior. Furthermore, the effects of polycyclic aromatic hydrocarbons (PAHs), major components of PM, on VSMC migration and the underlying mechanisms were examined. RESULTS: VSMC migration was significantly increased by treatment with organic matters extracted from ambient PM. The total amount of PAHs contained in WPM was higher than that in SPM, leading to higher ROS generation and VSMC migration. The increased migration was successfully inhibited by treatment with the anti-oxidant, N-acetyl-cysteine (NAC). The levels of matrix metalloproteinase (MMP) 2 and 9 were significantly increased in ambient PM-treated VSMCs, with MMP9 levels being significantly higher in WPM-treated VSMCs than in those treated with SPM. As expected, migration was significantly increased in all tested PAHs (anthracene, ANT; benz(a)anthracene, BaA) and their oxygenated derivatives (9,10-Anthraquinone, AQ; 7,12-benz(a)anthraquinone, BAQ, respectively). The phosphorylated levels of focal adhesion kinase (FAK) and formation of the focal adhesion complex were significantly increased in ambient PM or PAH-treated VSMCs, and these effects were blocked by administration of NAC or α-NF, an inhibitor of AhR, the receptor that allows PAH uptake. Subsequently, the levels of phosphorylated Src and NRF, the downstream targets of FAK, were altered with a pattern similar to that of p-FAK. CONCLUSIONS: PAHs, including oxy-PAHs, in ambient PM may have dual effects that lead to an increase in VSMC migration. One is the generation of oxidative stress followed by MMP upregulation, and the other is actin reorganization that results from the activation of the focal adhesion complex.


Subject(s)
Air Pollutants , Atherosclerosis , Polycyclic Aromatic Hydrocarbons , Actins , Air Pollutants/analysis , Cell Movement , Humans , Matrix Metalloproteinases , Muscle, Smooth, Vascular/chemistry , Oxidative Stress , Particulate Matter/analysis , Particulate Matter/toxicity , Polycyclic Aromatic Hydrocarbons/analysis , Polycyclic Aromatic Hydrocarbons/toxicity , Up-Regulation
4.
Arch Physiol Biochem ; 128(2): 490-500, 2022 Apr.
Article in English | MEDLINE | ID: mdl-31794282

ABSTRACT

OBJECTIVE: This study was to investigate the underlying mechanisms of valsartan chronotherapy in regulating blood pressure variability. METHODS: RT-PCR was used to assay clock genes expression rhythm in the hypothalamus, aortic vessels, and target organs after valsartan chronotherapy. WB was used to measure Period 1 (Per1), Period 2 (Per2) protein expression in aortic vessels, as well as to measure phosphorylation of 20-kDa regulatory myosin light chain (MLC20) in VSMCs. RESULTS: Specific clock genes in the hypothalamus, and Per1 and Per2 in aorta abdominalis, exhibited disordered circadian expression in vivo. Valsartan asleep time administration (VSA) restored circadian clock gene expression in a tissue- and gene-specific manner. In vitro, VSA was more efficient in blocking angiotensin II relative to VWA, which led to differential circadian rhythms of Per1 and Per2, ultimately corrected MLC20 phosphorylation. CONCLUSION: VSA may be efficacious in regulating circadian clock genes rhythm, then concomitantly correct circadian blood pressure rhythms.


Subject(s)
Chronotherapy , Muscle, Smooth, Vascular , Animals , Gene Expression , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , Rats , Rats, Inbred SHR , Valsartan/pharmacology
5.
J Clin Lab Anal ; 35(8): e23854, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34313357

ABSTRACT

OBJECTIVE: In order to find new strategies for the prevention of vascular calcification in uremic individuals especially treated by dialysis and develop novel therapeutic targets in vascular calcification, we explore the role of KCa3.1 in alkalinization-induced VSMCs calcification in vitro. METHOD: Rat VSMCs calcification model was established by beta-glycerophosphate (ß-GP, 10 mM) induction. The pH of Dulbecco's modified Eagle's medium (DMEM) was adjusted every 24 h with 10 mM HCl or 10 mM NaHCO3 . The mineralization was measured by Alizarin Red staining and O-cresolphthalein complex one method. mRNA and protein expression were detected by RT-PCR and Western blot or immunofluorescence. Ca2+ influx was measured by Elisa. RESULT: The results indicated that alkalization induced an increase in Ca2+ influx to enhance VSMCs calcification. Furthermore, the increase of calcification was associated with the expression of KCa3.1 via advanced expression of osteoblastic differentiation markers alkaline phosphatase (ALP) and Runt-related transcription factor 2 (Runx2). Blocking KCa3.1 with TRAM-34 or shRNA vector can significantly lowered the effects of calcification in the activity of ALP and Runx2 expression. CONCLUSION: Together all, our studies suggested that alkalinization can promote vascular calcification by upregulating KCa3.1 channel and enhancing osteogenic/chondrogenic differentiation by upregulating Runx2. The specific inhibitor TRAM-34 and KCa3.1-shRNA ameliorated VSMCs calcification by downregulating KCa3.1.


Subject(s)
Aorta/pathology , Calcinosis/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Alkaline Phosphatase/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Calcinosis/chemically induced , Calcinosis/drug therapy , Calcium/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Glycerophosphates/toxicity , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pyrazoles/pharmacology , Rats, Sprague-Dawley
6.
Environ Sci Technol ; 54(22): 14482-14493, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33138382

ABSTRACT

Coal combustion emits a large amount of PM2.5 (particulate matters with aerodynamic diameters less than 2.5 µm) and causes adverse damages to the cardiovascular system. In this study, emissions from anthracite and bitumite were examined. Red mud (RM) acts as an additive and is mixed in coal briquettes with a content of 0-10% as a single variable to demonstrate the reduction in the PM2.5 emissions. Burnt in a regulated combustion chamber, the 10% RM-containing bitumite and anthracite briquettes showed 52.3 and 18.6% reduction in PM2.5, respectively, compared with their chunk coals. Lower cytotoxicity (in terms of oxidative stresses and inflammation factors) was observed for PM2.5 emitted from the RM-containing briquettes than those from non-RM briquettes, especially for the bitumite groups. Besides, the results of western blotting illustrated that the inhibition of NF-κB and MAPK was the potential pathway for the reduction of cytokine levels by the RM addition. The regression analyses further demonstrated that the reduction was attributed to the lower emissions of transition metals (i.e., Mn) and PAHs (i.e., acenaphthene). This pilot study provides solid evidence for the cytotoxicity to vascular smooth muscle cells induced by PM2.5 from coal combustion and potential solutions for reducing the emission of toxic pollutants from human health perspectives.


Subject(s)
Air Pollutants , Coal , Air Pollutants/analysis , China , Coal/analysis , Environmental Monitoring , Humans , Muscle, Smooth, Vascular/chemistry , Particulate Matter/analysis , Pilot Projects
7.
Life Sci ; 260: 118280, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32800835

ABSTRACT

AIMS: Vascular calcification is a common complication in patients with chronic kidney disease and associated with increased morbidity and mortality. The role of TRPM7 in vascular smooth muscle cell (VSMC) transformation during vascular calcification is not clear. We aim to investigate the effects of phosphate and indoxyl sulphate on the expression of TRPM7 and calcification-related molecules in VSMC. MAIN METHODS: Human aortic smooth muscle cells (HASMC) were treated with phosphate (3.3 mM) or indoxyl sulphate (500 µM and 1000 µM). 2-APB, a channel blocker of TRPM7 was added simultaneously in blocking experiment. Cells were then examined grossly and alizarin red solution was employed for calcification assessment. Lastly, cells were harvested for gene expression and protein abundance analysis. KEY FINDINGS: Phosphate treatment induced significant increase in BMP2, RUNX2, BMP7, vitamin D receptor (VDR), calcium sensing receptor (CaSR) and TRPM7, but 1-alpha hydroxylase, klotho, DKK1 and sclerostin were not changed. The addition of 2-APB prevented increase of BMP2, RUNX2, BMP7, VDR, CaSR and TRPM7. Indoxyl sulphate treatment was associated with decrease in TRPM7 and DKK1, but increase in RUNX2, BMP2 and VDR were noted. There were no significant alterations in BMP7, CaSR, klotho,1-alpha hydroxylase and sclerostin. Co-treatment with 2-APB reversed the increase in VDR. SIGNIFICANCE: Both phosphate and indoxyl sulphate induced calcification in VSMC but it was more prominent in phosphate. TRPM7 was upregulated by phosphate but downregulated in indoxyl sulphate treatment. Vascular calcification was reduced by blocking TRPM7 with 2-APB and there was partial anti-calcification effect in indoxyl sulphate.


Subject(s)
Indican/pharmacology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Phosphates/pharmacology , Protein Serine-Threonine Kinases/physiology , TRPM Cation Channels/physiology , Vascular Calcification/physiopathology , Bone Morphogenetic Protein 2/analysis , Bone Morphogenetic Protein 7/analysis , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/analysis , Humans , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/drug effects , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Calcitriol/analysis , Renal Insufficiency, Chronic/complications , TRPM Cation Channels/analysis , TRPM Cation Channels/antagonists & inhibitors , Vascular Calcification/chemically induced , Vascular Calcification/etiology
8.
Arterioscler Thromb Vasc Biol ; 40(6): e166-e179, 2020 06.
Article in English | MEDLINE | ID: mdl-32349534

ABSTRACT

OBJECTIVE: Recent studies suggest that the P2Y12 (P2Y purinoceptor 12) receptor of vascular smooth muscle cells in atherosclerotic plaques aggravates atherosclerosis, and P2Y12 receptor inhibitors such as CDL (clopidogrel) may effectively treat atherosclerosis. It is imperative to identify an effective biomarker for reflecting the P2Y12 receptor expression on vascular smooth muscle cells in plaques. Approach and Results: We found that there was a positive correlation between the level of circulating sLRP1 (soluble low-density lipoprotein receptor-related protein 1) and the number of LRP1+ α-SMA+ (α-smooth muscle actin), P2Y12+, or P2Y12+ LRP1+ cells in plaques from apoE-/- mice fed a high-fat diet. Furthermore, activation of the P2Y12 receptor increased the expression and shedding of LRP1 in vascular smooth muscle cells by inhibiting cAMP (3'-5'-cyclic adenosine monophosphate)/PKA (protein kinase A)/SREBP-2 (sterol regulatory element binding transcription factor 2). Conversely, genetic knockdown or pharmacological inhibition of the P2Y12 receptor had the opposite effects. Additionally, CDL decreased the number of lesional LRP1+ α-SMA+ cells and the levels of circulating sLRP1 by activating cAMP/PKA/SREBP-2 in apoE-/- mice fed a high-fat diet. CONCLUSIONS: Our study suggests that sLRP1 may be a biomarker that reflects the P2Y12 receptor level in plaques and has the potential to be an indicator for administering P2Y12 receptor inhibitors for patients with atherosclerosis.


Subject(s)
Biomarkers/analysis , Gene Expression , Low Density Lipoprotein Receptor-Related Protein-1/analysis , Plaque, Atherosclerotic/metabolism , Receptors, Purinergic P2Y12/genetics , Actins/analysis , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Clopidogrel/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat , Gene Knockdown Techniques , Low Density Lipoprotein Receptor-Related Protein-1/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/metabolism , Plaque, Atherosclerotic/chemistry , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/physiology , Signal Transduction , Sterol Regulatory Element Binding Protein 2/metabolism
9.
Arterioscler Thromb Vasc Biol ; 40(6): 1559-1573, 2020 06.
Article in English | MEDLINE | ID: mdl-32321307

ABSTRACT

OBJECTIVE: Excessive prostaglandin E2 production is a hallmark of abdominal aortic aneurysm (AAA). Enhanced expression of prostaglandin E2 receptor EP4 (prostaglandin E receptor 4) in vascular smooth muscle cells (VSMCs) has been demonstrated in human AAAs. Although moderate expression of EP4 contributes to vascular homeostasis, the roles of excessive EP4 in vascular pathology remain uncertain. We aimed to investigate whether EP4 overexpression in VSMCs exacerbates AAAs. Approach and Results: We constructed mice with EP4 overexpressed selectively in VSMCs under an SM22α promoter (EP4-Tg). Most EP4-Tg mice died within 2 weeks of Ang II (angiotensin II) infusion due to AAA, while nontransgenic mice given Ang II displayed no overt phenotype. EP4-Tg developed much larger AAAs than nontransgenic mice after periaortic CaCl2 application. In contrast, EP4fl/+;SM22-Cre;ApoE-/- and EP4fl/+;SM22-Cre mice, which are EP4 heterozygous knockout in VSMCs, rarely exhibited AAA after Ang II or CaCl2 treatment, respectively. In Ang II-infused EP4-Tg aorta, Ly6Chi inflammatory monocyte/macrophage infiltration and MMP-9 (matrix metalloprotease-9) activation were enhanced. An unbiased analysis revealed that EP4 stimulation positively regulated the genes binding cytokine receptors in VSMCs, in which IL (interleukin)-6 was the most strongly upregulated. In VSMCs of EP4-Tg and human AAAs, EP4 stimulation caused marked IL-6 production via TAK1 (transforming growth factor-ß-activated kinase 1), NF-κB (nuclear factor-kappa B), JNK (c-Jun N-terminal kinase), and p38. Inhibition of IL-6 prevented Ang II-induced AAA formation in EP4-Tg. In addition, EP4 stimulation decreased elastin/collagen cross-linking protein LOX (lysyl oxidase) in both human and mouse VSMCs. CONCLUSIONS: Dysregulated EP4 overexpression in VSMCs promotes inflammatory monocyte/macrophage infiltration and attenuates elastin/collagen fiber formation, leading to AAA exacerbation.


Subject(s)
Aortic Aneurysm, Abdominal/etiology , Inflammation/etiology , Muscle, Smooth, Vascular/metabolism , Receptors, Prostaglandin E, EP4 Subtype/physiology , Signal Transduction/physiology , Angiotensin II/administration & dosage , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Calcium Chloride/administration & dosage , Gene Expression , Gene Expression Regulation/physiology , Humans , Interleukin-6/genetics , Macrophages/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Mice, Transgenic , Monocytes/pathology , Muscle, Smooth, Vascular/chemistry , Myocytes, Smooth Muscle/metabolism , Protein-Lysine 6-Oxidase/analysis , Protein-Lysine 6-Oxidase/genetics , Receptors, Cytokine/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics
10.
J Biochem ; 166(5): 383-392, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31504625

ABSTRACT

The apelin receptor (APJ), a receptor for apelin and elabela/apela, induces vasodilation and vasoconstriction in blood vessels. However, the prolonged effects of increased APJ-mediated signalling, involving vasoconstriction, in smooth muscle cells have not been fully characterized. Here, we investigated the vasoactive effects of APJ gain of function under the control of the smooth muscle actin (SMA) gene promoter in mice. Transgenic overexpression of APJ (SMA-APJ) conferred sensitivity to blood pressure and vascular contraction induced by apelin administration in vivo. Interestingly, ex vivo experiments showed that apelin markedly increased the vasoconstriction of isolated aorta induced by noradrenaline (NA), an agonist for α- and ß-adrenergic receptors, or phenylephrine, a specific agonist for α1-adrenergic receptor (α1-AR). In addition, intracellular calcium influx was augmented by apelin with NA in HEK293T cells expressing APJ and α1A-AR. To examine the cooperative action of APJ and α1A-AR in the regulation of vasoconstriction, we developed α1A-AR deficient mice using a genome-editing technique, and then established SMA-APJ/α1A-AR-KO mice. In the latter mouse line, aortic vasoconstriction induced by a specific agonist for α1A-AR, A-61603, were significantly less than in SMA-APJ mice. These results suggest that the APJ-enhanced response requires α1A-AR to contract vessels coordinately.


Subject(s)
Apelin Receptors/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Vasoconstriction , Animals , Humans , Mice , Mice, Inbred ICR , Mice, Transgenic , Muscle, Smooth, Vascular/chemistry
11.
Cells ; 8(8)2019 07 31.
Article in English | MEDLINE | ID: mdl-31370272

ABSTRACT

Regulation of vascular smooth muscle cell (VSMC) proliferation is essential to maintain vascular homeostasis. Hypoxia induces abnormal proliferation of VSMCs and causes vascular proliferative disorders, such as pulmonary hypertension and atherosclerosis. As several cyclin/cyclin-dependent kinase (CDK) complexes and CDK inhibitors (CKIs) control cell proliferation, in this study, we investigated CKIs involved in the hypoxia-induced proliferation process of human primary pulmonary artery smooth muscle cells to understand the underlying molecular mechanism. We demonstrated that p15, p16, and p21 are downregulated in pulmonary artery smooth muscle cells when exposed to hypoxia. In addition, we identified novel hypoxia-induced microRNAs (hypoxamiRs) including miR-497, miR-1268a, and miR-665 that are upregulated under hypoxia and post-transcriptionally regulate p15, p16, and p21 genes, respectively, by directly targeting their 3'UTRs. These miRNAs promoted the proliferation of VSMCs, and their inhibition decreased VSMC proliferation even in hypoxic conditions. Overall, this study revealed that miRNA-mediated regulatory mechanism of CKIs is essential for hypoxia-induced proliferation of VSMCs. These findings provide insights for a better understanding of the pathogenesis of vascular proliferative disorders.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Down-Regulation , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Pulmonary Artery/cytology , 3' Untranslated Regions , Animals , COS Cells , Cell Hypoxia , Cell Line , Cell Proliferation , Chlorocebus aethiops , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Homeostasis , Humans , Muscle, Smooth, Vascular/chemistry , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/chemistry
12.
Clin Exp Nephrol ; 23(9): 1100-1108, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31214872

ABSTRACT

BACKGROUND: Patients with uremia have an excessive mortality from cardiovascular disease (CVD). Arterial remodeling is mainly responsible for uremia-induced CVD and has been well studied, yet venous remodeling is poorly understood. Here we investigate the histopathology and proteomic profiles of venous remodeling in uremic patients. METHODS: Forearm cephalic veins were isolated from nine uremic patients during surgeries for arteriovenous fistula, and from nine healthy controls when applying surgical debridement. Hematoxylin-eosin, Masson's trichrome, von Kossa, and immunohistochemistry (IHC) against proliferating cell nuclear antigen were stained for histopathology. Isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis was executed to explore the proteome of the veins. The core regulatory protein was validated by western blot, IHC, and immunofluorescence. RESULTS: Phlebosclerosis, characterized by intimal rarefaction and medial thickening with disordered proliferation of vascular smooth muscle cells (VSMCs), was the prominent pathological manifestation of peripheral veins in uremic patients, while inflammatory cell infiltration, atherosclerosis or calcification were not obviously detected. iTRAQ analysis showed that 350 proteins were significantly changed in phlebosclerosis of uremic patients compared with healthy controls, of which integrin-ß1 (ITGß1) exhibited the strongest regulatory ability by intermolecular interaction network analysis. The enhanced ITGß1 expression was mainly co-expressed with the disordered proliferation of VSMCs while a little with vascular endothelial cells in the forearm cephalic veins of uremic patients. CONCLUSIONS: Phlebosclerosis is the prominent pathological manifestation in peripheral veins of uremic patients. This pathological alteration mainly attributes to the disordered proliferation of VSMCs, which is potentially mediated by ITGß1.


Subject(s)
Forearm/blood supply , Integrin beta1/analysis , Peripheral Vascular Diseases/etiology , Proteomics/methods , Uremia/complications , Vascular Remodeling , Veins/chemistry , Veins/pathology , Case-Control Studies , Cell Proliferation , Endothelial Cells/chemistry , Endothelial Cells/pathology , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/pathology , Peripheral Vascular Diseases/metabolism , Peripheral Vascular Diseases/pathology , Sclerosis , Uremia/diagnosis
13.
J Cereb Blood Flow Metab ; 39(7): 1369-1380, 2019 07.
Article in English | MEDLINE | ID: mdl-29480757

ABSTRACT

Platelet-derived growth factor receptor-ß (PDGFR-ß) has been reported to promote phenotypic transformation of vascular smooth muscle cells (VSMCs). The purpose of this study was to investigate the role of the PDGFR-ß/IRF9/SIRT-1/NF-κB pathway in VSMC phenotypic transformation after subarachnoid hemorrhage (SAH). SAH was induced using the endovascular perforation model in Sprague-Dawley rats. PDGFR-ß small interfering RNA (siRNA) and IRF9 siRNA were injected intracerebroventricularly 48 h before SAH. SIRT1 activator (resveratrol) and inhibitor (EX527) were administered intraperitoneally 1 h after SAH induction. Twenty-four hours after SAH, the VSMC contractile phenotype marker α-smooth muscle actin (α-SMA) decreased, whereas the VSMC synthetic phenotype marker embryonic smooth muscle myosin heavy chain (Smemb) increased. Both PDGFR-ß siRNA and IRF9 siRNA attenuated the induction of nuclear factor-κB (NF-κB) and enhanced the expression of α-SMA. The SIRT1 activator (resveratrol) preserved VSMC contractile phenotype, significantly alleviated neurological dysfunction, and reduced brain edema. However, these beneficial effects of PDGFR-ß siRNA, IRF9 siRNA and resveratrol were abolished by the SIRT1 inhibitor (EX527). This study shows that PDGFR-ß/IRF9/SIRT-1/NF-κB signaling played a role in the VSMC phenotypic transformation after SAH. Inhibition of this signaling cascade preserved the contractile phenotype of VSMCs, thereby improving neurological outcomes following SAH.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/physiology , Muscle, Smooth, Vascular/physiopathology , NF-kappa B/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Sirtuin 1/physiology , Subarachnoid Hemorrhage/physiopathology , Actins/analysis , Animals , Disease Models, Animal , Gene Knockdown Techniques , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Male , Muscle, Smooth, Vascular/chemistry , Myosin Heavy Chains/analysis , Phenotype , RNA, Small Interfering/administration & dosage , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/genetics , Resveratrol/pharmacology , Signal Transduction/physiology , Sirtuin 1/antagonists & inhibitors , Subarachnoid Hemorrhage/etiology
14.
Chem Phys Lipids ; 218: 149-157, 2019 01.
Article in English | MEDLINE | ID: mdl-30582896

ABSTRACT

Liposomal delivery systems (LDSs) have been at the forefront of medicinal nanotechnology for over three decades. Increasing LDS association to target cells and cargo delivery is crucial to bolstering overall nanodrug efficacy. Our laboratory aims to develop LDSs for molecular therapeutics aimed at vascular pathology. We have previously established a liposome platform that is an effective delivery system for RNA interference in vascular cell types by using polyethylene glycol (PEG) decorated liposomes bearing an octa-arginine (R8) cell penetrating peptide (CPP). Further tailoring liposome membranes to mimic vascular cell membrane lipid constituents may be a promising strategy for increasing cargo delivery. Here we aimed to develop liposomal formulations that could make use of diacylglycerol (DAG) and phosphatidylserine (PS), naturally occurring lipid species that are known to influence vascular cell function, as a facile and efficient means to increase nanodrug efficacy without compromising clinical viability. We investigated the ability of DAG and PS to amplify the cellular uptake of our previously established LDS platform loaded with small interfering ribonucleic acid (siRNA) cargo. Cellular fluorescence microscopy experiments were performed in conjunction with quantitative cell association assays and cytotoxicity assays to analyze the effect of DAG/PS on the differential delivery of fluorescently-tagged liposomes to vascular smooth muscle cells (VSMCs) and vascular endothelial cells (VECs) and on liposomal-mediated toxicity. In these studies, significant, dose-dependent increases in association to target cells were observed, as well as cell-type specific effects on cell viability. The stability and encapsulation-efficiency of the DAG/PS-modified LDSs were analyzed by standard nanoparticle characterization methods, and siRNA transfection efficacy was quantified to gauge delivery potential as a function of DAG/PS modification. Our results suggest that the signaling lipids tested here imbue our LDS architectures with increased therapeutic potential, without compromising stability, encapsulation efficiency, or biocompatibility, thus presenting a natural strategy to increase nanodrug efficacy and specificity.


Subject(s)
Diglycerides/chemistry , Drug Delivery Systems , Endothelial Cells/chemistry , Muscle, Smooth, Vascular/chemistry , Nanoparticles/chemistry , Phosphatidylserines/chemistry , RNA, Small Interfering/pharmacology , Aorta/cytology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Humans , Liposomes/chemistry , Molecular Structure , RNA, Small Interfering/chemistry , Structure-Activity Relationship
15.
Anat Rec (Hoboken) ; 301(11): 1917-1927, 2018 11.
Article in English | MEDLINE | ID: mdl-30288932

ABSTRACT

It is well known that nerves modulate the development and remodeling of blood vessels by releasing different neuropeptides and neurotransmitters. Secretoneurin (SN), a neuropeptide located in nerve fibers along blood vessels, acts as a pro-angiogenic agent and induces postnatal vasculogenesis. However, little is known about its involvement in arteriogenesis. In the present study, we tested the hypothesis that SN promotes arteriogenesis in a rat model of hind limb ischemia, as such, we evaluated the effect of this neuropeptide on proliferation and the production of adhesion and chemotaxis molecules in vascular smooth muscle cells (VSMCs), the main component that carries the burden of the transformation of a small arteriole into a large collateral vessel. In vivo, SN-immunoreactive nerve fibers were abundantly distributed in the adventitia of the collateral vessel. Moreover, administration of SN induced cell proliferation in the vascular wall and the infiltration of inflammatory cells/macrophages to promote collateral vessel growth. This was shown by an increased density of arterioles/arteries, together with a well-developed network of collateral vessels, and well-preserved skeletal muscles. In vitro, SN exerted proliferative effects on VSMCs and stimulated these cells to express adhesion molecules. In conclusion, our data demonstrate for the first time that SN acts as a mediator of inflammation, contributing to collateral vessel growth, in addition to directly stimulating cell proliferation in the vascular wall to promote collateral vessel growth in a rat model of hind limb ischemia. Anat Rec, 301:1917-1927, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Collateral Circulation/physiology , Femoral Artery/metabolism , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/physiology , Neuropeptides/metabolism , Secretogranin II/metabolism , Animals , Cells, Cultured , Femoral Artery/diagnostic imaging , Femoral Artery/drug effects , Hindlimb/blood supply , Hindlimb/diagnostic imaging , Hindlimb/metabolism , Ischemia/diagnostic imaging , Ischemia/metabolism , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Neuropeptides/pharmacology , Rats , Rats, Sprague-Dawley , Secretogranin II/pharmacology
16.
J Vasc Res ; 55(4): 224-234, 2018.
Article in English | MEDLINE | ID: mdl-30092589

ABSTRACT

Thyroid hormone (TH) regulates gene transcription by binding to TH receptors (TRs). TRs regulate the genes of lipid metabolism and the renin-angiotensin system (RAS). We examined the effect of TRα deletion in ApoE-/- mice (DKO mice) on the following: (i) the expression of genes controlling cholesterol metabolism and tissue (t)RAS in the liver and aorta and (ii) the expression of these genes and the regulation of cholesterol content in cultured vascular smooth muscle cells (VSMCs). TRα deletion in ApoE-/- mice led to the repression of genes involved in the synthesis and influx of cholesterol in the liver. However, TRα deletion in the arterial wall suppressed the expression of genes involved in the esterification and excretion of cholesterol and enhanced the expression of angiotensinogen (AGT). The VSMCs of the ApoE-/- and DKO mice increased their cholesterol content during cholesterol loading, but failed to increase the expression of ATP-binding cassette transporter A1 (ABCA1). T3 addition partially corrected these abnormalities in the cells of the ApoE-/- mice but not those of the DKO mice. In conclusion, TRα deletion in ApoE-/- mice slightly increases the expression of tRAS in the aorta and aggravates the dysregulation of cholesterol content in the VSMCs.


Subject(s)
Apolipoproteins E/deficiency , Cholesterol/metabolism , Muscle, Smooth, Vascular/metabolism , Renin-Angiotensin System/physiology , Thyroid Hormone Receptors alpha/deficiency , ATP Binding Cassette Transporter 1/genetics , Animals , Aorta/chemistry , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Atherosclerosis/diagnostic imaging , Cells, Cultured , Cholesterol/administration & dosage , Cholesterol/genetics , Gene Expression , Hybridization, Genetic , Liver/chemistry , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , RNA, Messenger , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/physiology , Triiodothyronine/pharmacology , Ultrasonography
17.
Arterioscler Thromb Vasc Biol ; 38(9): 2184-2190, 2018 09.
Article in English | MEDLINE | ID: mdl-29976770

ABSTRACT

Objective- Unreliable antibodies often hinder the accurate detection of an endogenous protein, and this is particularly true for the cardiac and smooth muscle cofactor, MYOCD (myocardin). Accordingly, the mouse Myocd locus was targeted with 2 independent epitope tags for the unambiguous expression, localization, and activity of MYOCD protein. Approach and Results- 3cCRISPR (3-component clustered regularly interspaced short palindromic repeat) was used to engineer a carboxyl-terminal 3×FLAG or 3×HA epitope tag in mouse embryos. Western blotting with antibodies to each tag revealed a MYOCD protein product of ≈150 kDa, a size considerably larger than that reported in virtually all publications. MYOCD protein was most abundant in some adult smooth muscle-containing tissues with surprisingly low-level expression in the heart. Both alleles of Myocd are active in aorta because a 2-fold increase in protein was seen in mice homozygous versus heterozygous for FLAG-tagged Myocd. ChIP (chromatin immunoprecipitation)-quantitative polymerase chain reaction studies provide proof-of-principle data demonstrating the utility of this mouse line in conducting genome-wide ChIP-seq studies to ascertain the full complement of MYOCD-dependent target genes in vivo. Although FLAG-tagged MYOCD protein was undetectable in sections of adult mouse tissues, low-passaged vascular smooth muscle cells exhibited expected nuclear localization. Conclusions- This report validates new mouse models for analyzing MYOCD protein expression, localization, and binding activity in vivo and highlights the need for rigorous authentication of antibodies in biomedical research.


Subject(s)
CRISPR-Cas Systems , Epitope Mapping/methods , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Trans-Activators/analysis , Trans-Activators/metabolism , Animals , Embryo, Mammalian , Epitopes/analysis , Mice , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/chemistry
18.
Nephrology (Carlton) ; 23 Suppl 2: 58-62, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29968404

ABSTRACT

AIM: The present study was performed to examine the clinicopathological significance of hyaline deposits in the smooth muscle of the interlobular artery (interlobular hyaline arteriopathy [IHA]) in renal allografts. METHODS: Tissue specimens that included the interlobular artery from biopsies performed from January 2012 to December 2015, as well as specimens from biopsies performed ≥1 year after living kidney transplantation were analyzed. Biopsies of recipients with new-onset diabetes mellitus after transplantation were excluded, as well as those of recipients who had undergone transplantation because of diabetic nephropathy. Arteriolopathy was evaluated using the aah score determined by the Banff 2007 classification. RESULTS: In total, 51 specimens with IHA lesions were identified among 381 biopsies obtained from 243 recipients performed ≥1 year after kidney transplantation. Among these 51 biopsies, 18 specimens had a score of aah3, 29 had a score of aah2, and four had a score of aah1. The incidence of IHA lesions was 3.6% at ≥1 to <4 years, 18.5% at ≥4 to <8 years, and 54.1% at ≥8 years. Older kidney grafts exhibited more IHA lesions. Among the biopsy specimens obtained ≥8 years after transplantation, no significant differences in the recipient or donor age, duration after transplantation, or prevalence of hypertension were observed between the IHA and non-IHA groups. The aah scores were significantly higher in the IHA group ≥8 years after transplantation as determined by the mean score test (P < 0.01). CONCLUSION: IHA in renal allografts is associated with severe arteriolopathy.


Subject(s)
Hyalin , Kidney Transplantation/adverse effects , Kidney/blood supply , Muscle, Smooth, Vascular/chemistry , Vascular Diseases/metabolism , Allografts , Arterioles/chemistry , Arterioles/pathology , Biopsy , Humans , Incidence , Kidney Transplantation/methods , Living Donors , Muscle, Smooth, Vascular/pathology , Prevalence , Renal Artery/chemistry , Renal Artery/pathology , Severity of Illness Index , Time Factors , Tokyo/epidemiology , Treatment Outcome , Vascular Diseases/epidemiology , Vascular Diseases/pathology
19.
Neuron ; 99(2): 362-375.e4, 2018 07 25.
Article in English | MEDLINE | ID: mdl-29937277

ABSTRACT

Functional hyperemia, a regional increase of blood flow triggered by local neural activation, is used to map brain activity in health and disease. However, the spatial-temporal dynamics of functional hyperemia remain unclear. Two-photon imaging of the entire vascular arbor in NG2-creERT2;GCaMP6f mice shows that local synaptic activation, measured via oligodendrocyte precursor cell (OPC) Ca2+ signaling, generates a synchronous Ca2+ drop in pericytes and smooth muscle cells (SMCs) enwrapping all upstream vessels feeding the activated synapses. Surprisingly, the onset timing, direction, and amplitude of vessel diameter and blood velocity changes vary dramatically from juxta-synaptic capillaries back to the pial arteriole. These results establish a precise spatial-temporal sequence of vascular changes triggered by neural activity and essential for the interpretation of blood-flow-based imaging techniques such as BOLD-fMRI.


Subject(s)
Brain/blood supply , Brain/physiology , Hyperemia/physiopathology , Pia Mater/blood supply , Pia Mater/physiology , Synapses/physiology , Animals , Brain Chemistry/physiology , Hyperemia/diagnosis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal/methods , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/physiology , Pericytes/chemistry , Pericytes/physiology , Pia Mater/chemistry , Synapses/chemistry
20.
Atherosclerosis ; 271: 237-244, 2018 04.
Article in English | MEDLINE | ID: mdl-29395098

ABSTRACT

BACKGROUND AND AIMS: We aim to identify significant transcriptome alterations of vascular smooth muscle cells (VSMCs) in the aortic wall of myocardial infarction (MI) patients. Providing a robust transcriptomic signature, we aim to highlight the most likely aberrant pathway(s) in MI VSMCs. METHODS AND RESULTS: Laser-captured microdissection (LCM) was used to obtain VSMCs from aortic wall tissues harvested during coronary artery bypass surgery. Microarray gene analysis was applied to analyse VSMCs from 17 MI and 19 non-MI patients. Prediction Analysis of Microarray (PAM) identified 370 genes that significantly discriminated MI and non-MI samples and were enriched in genes responsible for muscle development, differentiation and phenotype regulation. Incorporation of gene ontology (GO) led to the identification of a 21-gene VSMCs-associated classifier that discriminated between MI and non-MI patients with 92% accuracy. The mass spectrometry-based iTRAQ analysis of the MI and non-MI samples revealed 94 proteins significantly differentiating these tissues. Ingenuity Pathway Analysis (IPA) of 370 genes revealed top pathways associated with hypoxia signaling in the cardiovascular system. Enrichment analysis of these proteins suggested an activation of the superoxide radical degradation pathway. An integrated transcriptome-proteome pathway analysis revealed that superoxide radical degradation pathway remained the most implicated pathway. The intersection of the top candidate molecules from the transcriptome and proteome highlighted superoxide dismutase (SOD1) overexpression. CONCLUSIONS: We provided a novel 21-gene VSMCs-associated MI classifier in reference to significant VSMCs transcriptome alterations that, in combination with proteomics data, suggests the activation of superoxide radical degradation pathway in VSMCs of MI patients.


Subject(s)
Muscle, Smooth, Vascular/chemistry , Myocardial Infarction/genetics , Myocytes, Smooth Muscle/chemistry , Signal Transduction/genetics , Transcriptome , Aorta/chemistry , Case-Control Studies , Chromatography, Liquid , Gene Expression Profiling/methods , Humans , Myocardial Infarction/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proteomics/methods , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Superoxides/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...