Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 963
Filter
Add more filters

Publication year range
1.
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
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.
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
4.
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
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.
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
8.
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
9.
Biochim Biophys Acta Mol Cell Res ; 1864(2): 336-344, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27888098

ABSTRACT

Maspin is a non-inhibitory member of the serpin family that affects cell behaviours related to migration and survival. We have previously shown that peptides of the isolated G α-helix (G-helix) domain of maspin show bioactivity. Migration, invasion, adhesion and proliferation of vascular smooth muscle cells (VSMC) are important processes that contribute to the build-up of atherosclerotic plaques. Here we report the use of functional assays of these behaviours to investigate whether other maspin-derived peptides impact directly on VSMC; focusing on potential anti-atherogenic properties. We designed 18 new peptides from the structural moieties of maspin above ten amino acid residues in length and considered them beside the existing G-helix peptides. Of the novel peptides screened those with the sequences of maspin strand 4 and 5 of beta sheet B (S4B and S5B) reduced VSMC migration, invasion and proliferation, as well as increasing cell adhesion. A longer peptide combining these consecutive sequences showed a potentiation of responses, and a 7-mer contained all essential elements for functionality. This is the first time that these parts of maspin have been highlighted as having key roles affecting cell function. We present evidence for a mechanism whereby S4B and S5B act through ERK1/2 and AMP-activated protein kinase (AMPK) to influence VSMC responses.


Subject(s)
Muscle, Smooth, Vascular/physiology , Peptide Fragments/physiology , Serpins/physiology , Amino Acid Sequence , Cell Proliferation/physiology , Cells, Cultured , Humans , Integrin beta1/physiology , MAP Kinase Signaling System , Muscle, Smooth, Vascular/chemistry , Serpins/chemistry , Signal Transduction
10.
Microcirculation ; 25(1)2018 01.
Article in English | MEDLINE | ID: mdl-29072364

ABSTRACT

Voltage-gated K+ (Kv ) channels are major determinants of membrane potential in vascular smooth muscle cells (VSMCs) and regulate the diameter of small cerebral arteries and arterioles. However, the intracellular structures that govern the expression and function of vascular Kv channels are poorly understood. Scaffolding proteins including postsynaptic density 95 (PSD95) recently were identified in rat cerebral VSMCs. Primarily characterized in neurons, the PSD95 scaffold has more than 50 known binding partners, and it can mediate macromolecular signaling between cell-surface receptors and ion channels. In cerebral arteries, Shaker-type Kv 1 channels appear to associate with the PSD95 molecular scaffold, and PSD95 is required for the normal expression and vasodilator influence of members of this K+ channel gene family. Furthermore, recent findings suggest that the ß1-subtype adrenergic receptor is expressed in cerebral VSMCs and forms a functional vasodilator complex with Kv 1 channels on the PSD95 scaffold. Activation of ß1-subtype adrenergic receptors in VSMCs enables protein kinase A-dependent phosphorylation and opening of Kv 1 channels in the PSD95 complex; the subsequent K+ efflux mediates membrane hyperpolarization and vasodilation of small cerebral arteries. Early evidence from other studies suggests that other families of Kv channels and scaffolding proteins are expressed in VSMCs. Future investigations into these macromolecular complexes that modulate the expression and function of Kv channels may reveal unknown signaling cascades that regulate VSMC excitability and provide novel targets for ion channel-based medications to optimize vascular tone.


Subject(s)
Cerebrovascular Circulation , Disks Large Homolog 4 Protein/physiology , Potassium Channels, Voltage-Gated/metabolism , Receptors, Adrenergic, beta/physiology , Animals , Disks Large Homolog 4 Protein/metabolism , Humans , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Rats , Receptors, Adrenergic, beta/metabolism
11.
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
12.
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
13.
J Vasc Surg ; 66(5): 1553-1564.e6, 2017 11.
Article in English | MEDLINE | ID: mdl-27720318

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a frequent, potentially life-threatening, disease that can only be treated by surgical means such as open surgery or endovascular repair. No alternative treatment is currently available, and despite expanding knowledge about the pathomechanism, clinical trials on medical aneurysm abrogation have led to inconclusive results. The heterogeneity of human AAA based on histologic examination is thereby generally neglected. In this study we aimed to further elucidate the role of these differences in aneurysm disease. METHODS: Tissue samples from AAA and popliteal artery aneurysm patients were examined by histomorphologic analysis, immunohistochemistry, Western blot, and polymerase chain reaction. The results were correlated with clinical data such as aneurysm diameter and laboratory results. RESULTS: The morphology of human AAA vessel wall probes varies tremendously based on the grade of inflammation. This correlates with increasing intima/media thickness and upregulation of the vascular endothelial growth factor cascade but not with any clinical parameter or the aneurysm diameter. The phenotypic switch of vascular smooth muscle cells occurred regardless of the inflammatory state and expressional changes of the transcription factors Kruppel-like factor-4 and transforming growth factor-ß lead to differential protein localization in aneurysmal compared with control arteries. These changes were in similar manner also observed in samples from popliteal artery aneurysms, which, however, showed a more homogenous phenotype. CONCLUSIONS: Heterogeneity of AAA vessel walls based on inflammatory morphology does not correlate with AAA diameter yet harbors specific implications for basic research and possible aneurysm detection.


Subject(s)
Aneurysm/pathology , Aortic Aneurysm, Abdominal/pathology , Cell Dedifferentiation , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Aneurysm/diagnostic imaging , Aneurysm/metabolism , Angiogenic Proteins/analysis , Aorta, Abdominal/chemistry , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/metabolism , Aortography/methods , Biomarkers/analysis , Computed Tomography Angiography , Dilatation, Pathologic , Extracellular Matrix/chemistry , Extracellular Matrix/pathology , Extracellular Matrix Proteins/analysis , Humans , Inflammation/diagnostic imaging , Inflammation/metabolism , Inflammation Mediators/analysis , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/analysis , Muscle, Smooth, Vascular/chemistry , Myocytes, Smooth Muscle/chemistry , Phenotype , Popliteal Artery/chemistry , Popliteal Artery/diagnostic imaging , Popliteal Artery/pathology , Transforming Growth Factor beta/analysis , Vascular Remodeling
14.
J Proteome Res ; 15(6): 1995-2007, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27146950

ABSTRACT

Human embryonic stem cells (hESCs) are promising in regenerative medicine (RM) due to their differentiation plasticity and proliferation potential. However, a major challenge in RM is the generation of a vascular system to support nutrient flow to newly synthesized tissues. Here we refined an existing method to generate tight vessels by differentiating hESCs in CD34(+) vascular progenitor cells using chemically defined media and growth conditions. We selectively purified these cells from CD34(-) outgrowth populations also formed. To analyze these differentiation processes, we compared the proteomes of the hESCs with those of the CD34(+) and CD34(-) populations using high resolution mass spectrometry, label-free quantification, and multivariate analysis. Eighteen protein markers validate the differentiated phenotypes in immunological assays; nine of these were also detected by proteomics and show statistically significant differential abundance. Another 225 proteins show differential abundance between the three cell types. Sixty-three of these have known functions in CD34(+) and CD34(-) cells. CD34(+) cells synthesize proteins implicated in endothelial cell differentiation and smooth muscle formation, which support the bipotent phenotype of these progenitor cells. CD34(-) cells are more heterogeneous synthesizing muscular/osteogenic/chondrogenic/adipogenic lineage markers. The remaining >150 differentially abundant proteins in CD34(+) or CD34(-) cells raise testable hypotheses for future studies to probe vascular morphogenesis.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Proteome/analysis , Stem Cells/cytology , Antigens, CD34 , Cells, Cultured , Culture Media/pharmacology , Endothelial Cells/chemistry , Endothelial Cells/cytology , Human Embryonic Stem Cells/chemistry , Humans , Mass Spectrometry , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Stem Cells/chemistry
15.
Heart Vessels ; 31(5): 795-806, 2016 May.
Article in English | MEDLINE | ID: mdl-26129868

ABSTRACT

Patients with bicuspid aortic valve (BAV) and patients with Marfan syndrome (MFS) are more prone to develop aortic dilation and dissection compared to persons with a tricuspid aortic valve (TAV). To elucidate potential common and distinct pathways of clinical relevance, we compared the histopathological substrates of aortopathy. Ascending aortic wall biopsies were divided in five groups: BAV (n = 36) and TAV (n = 23) without and with dilation and non-dilated MFS (n = 8). General histologic features, apoptosis, the expression of markers for vascular smooth muscle cell (VSMC) maturation, markers predictive for ascending aortic dilation in BAV, and expression of fibrillin-1 were investigated. Both MFS and BAV showed an altered distribution and decreased fibrillin-1 expression in the aorta and a significantly lower level of differentiated VSMC markers. Interestingly, markers predictive for aortic dilation in BAV were not expressed in the MFS aorta. The aorta in MFS was similar to the aorta in dilated TAV with regard to the presence of medial degeneration and apoptosis, while other markers for degeneration and aging like inflammation and progerin expression were low in MFS, comparable to BAV. Both MFS and BAV aortas have immature VSMCs, while MFS and TAV patients have a similar increased rate of medial degeneration. However, the mechanism leading to apoptosis is expected to be different, being fibrillin-1 mutation induced increased angiotensin-receptor-pathway signaling in MFS and cardiovascular aging and increased progerin in TAV. Our findings could explain why angiotensin inhibition is successful in MFS and less effective in TAV and BAV patients.


Subject(s)
Aorta/pathology , Aortic Aneurysm/etiology , Aortic Dissection/etiology , Aortic Valve/abnormalities , Heart Valve Diseases/complications , Marfan Syndrome/complications , Adult , Aged , Aged, 80 and over , Aortic Dissection/metabolism , Aortic Dissection/pathology , Aorta/chemistry , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Aortic Valve/pathology , Apoptosis , Bicuspid Aortic Valve Disease , Biomarkers/analysis , Biopsy , Dilatation, Pathologic , Female , Fibrillin-1/analysis , Heart Valve Diseases/pathology , Humans , Immunohistochemistry , Male , Marfan Syndrome/pathology , Middle Aged , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/pathology , Necrosis , Proto-Oncogene Proteins c-kit/analysis , Young Adult
16.
Beijing Da Xue Xue Bao Yi Xue Ban ; 48(5): 756-765, 2016 10 18.
Article in Zh | MEDLINE | ID: mdl-27752152

ABSTRACT

OBJECTIVE: To study the change of microRNA during the early stage of high phosphorus induced vascular smooth muscle cell (VSMC) calcification and its related mechanism. METHODS: The in vitro calcification model was created through stimulating VSMC cell line A7r5 with high Pi (2.6 mmol/L) for 7 d. The calcification was validated through ocresolphthalein complexone colorimetry to detect the cellular calcium content, real-time PCR to measure the calcification-related gene expression and alizarin red staining to observe the formation of calcium nodules. Based on the cell calcification model, microRNA microarray array was applied to screen the profiles of microRNA expression in VSMC following high Pi stimulation for different periods (0, 3 and 12 h). The array data were analyzed by TAM tool to explore the activated signaling pathway. RESULTS: The calcium content of A7r5 cells induced by high Pi was increased 9.6 times high as cells without Pi treatment (P<0.05). VSMC contractile phenotype genes (SM-α actin, SM22) were down-regulated (P<0.05), while calcification-related genes (BMP2, MSX2, Runx2) were up-regulated (P<0.05) in VSMC stimulated by high Pi. The calcium nodules were obviously formed in cells after 7 d high Pi treatment. In microarray experiment, 680 individual microRNAs were detected in high Pi-treated VSMCs at different time points (0, 3 and 12 h). Among these genes, miR-183, miR-664 and miR-9* were increased whereas miR-542-5P, let-7f and miR-29a were decreased in time-dependent manners. Twenty-six kinds of signaling pathways, including cell apoptosis, differentiation and proliferation, were significantly activated. All these activated pathways were associated with calcification. CONCLUSION: This study implies that microRNA changed in high Pi-induced VSMCs may involve in the process of calcification.


Subject(s)
Gene Expression Regulation/drug effects , MicroRNAs/pharmacology , MicroRNAs/physiology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/drug effects , Vascular Calcification/genetics , Vascular Calcification/physiopathology , Actins , Animals , Apoptosis/physiology , Bone Morphogenetic Protein 2/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Cells, Cultured/drug effects , Colorimetry , Core Binding Factor Alpha 1 Subunit , Down-Regulation , Gene Expression Regulation/genetics , Homeodomain Proteins , Microfilament Proteins , Muscle Proteins , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/physiology , Phosphorus/physiology , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation
17.
Circ Res ; 113(4): 365-71, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23801067

ABSTRACT

RATIONALE: Cyclic GMP (cGMP) is an important intracellular signaling molecule in the cardiovascular system, but its spatiotemporal dynamics in vivo is largely unknown. OBJECTIVE: To generate and characterize transgenic mice expressing the fluorescence resonance energy transfer-based ratiometric cGMP sensor, cGMP indicator with an EC50 of 500 nmol/L (cGi500), in cardiovascular tissues. METHODS AND RESULTS: Mouse lines with smooth muscle-specific or ubiquitous expression of cGi500 were generated by random transgenesis using an SM22α promoter fragment or by targeted integration of a Cre recombinase-activatable expression cassette driven by the cytomegalovirus early enhancer/chicken ß-actin/ß-globin promoter into the Rosa26 locus, respectively. Primary smooth muscle cells isolated from aorta, bladder, and colon of cGi500 mice showed strong sensor fluorescence. Basal cGMP concentrations were < 100 nmol/L, whereas stimulation with cGMP-elevating agents such as 2-(N,N-diethylamino)-diazenolate-2-oxide diethylammonium salt (DEA/NO) or the natriuretic peptides, atrial natriuretic peptide, and C-type natriuretic peptide evoked fluorescence resonance energy transfer changes corresponding to cGMP peak concentrations of ≈ 3 µmol/L. However, different types of smooth muscle cells had different sensitivities of their cGMP responses to DEA/NO, atrial natriuretic peptide, and C-type natriuretic peptide. Robust nitric oxide-induced cGMP transients with peak concentrations of ≈ 1 to > 3 µmol/L could also be monitored in blood vessels of the isolated retina and in the cremaster microcirculation of anesthetized mice. Moreover, with the use of a dorsal skinfold chamber model and multiphoton fluorescence resonance energy transfer microscopy, nitric oxide-stimulated vascular cGMP signals associated with vasodilation were detected in vivo in an acutely untouched preparation. CONCLUSIONS: These cGi500 transgenic mice permit the visualization of cardiovascular cGMP signals in live cells, tissues, and mice under normal and pathological conditions or during pharmacotherapy with cGMP-elevating drugs.


Subject(s)
Cardiovascular System/chemistry , Cyclic GMP/analysis , Cyclic GMP/genetics , Fluorescence Resonance Energy Transfer/methods , Mice, Transgenic/genetics , Signal Transduction/genetics , Animals , Biosensing Techniques/methods , Mice , Microscopy, Fluorescence, Multiphoton/methods , Models, Animal , Muscle, Smooth/chemistry , Muscle, Smooth, Vascular/chemistry
18.
J Surg Res ; 196(1): 180-9, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25801975

ABSTRACT

BACKGROUND: Nitric oxide (NO) more effectively inhibits neointimal hyperplasia in type 2 diabetic versus nondiabetic and type 1 diabetic rodents. NO also decreases the ubiquitin-conjugating enzyme UbcH10, which is critical to cell-cycle regulation. This study seeks to determine whether UbcH10 levels in the vasculature of diabetic animal models account for the differential efficacy of NO at inhibiting neointimal hyperplasia. MATERIALS AND METHODS: Vascular smooth muscle cells (VSMCs) harvested from nondiabetic lean Zucker (LZ) and type 2 diabetic Zucker diabetic fatty (ZDF) rats were exposed to high glucose (25 mM) and high insulin (24 nM) conditions to mimic the diabetic environment in vitro. LZ, streptozotocin-injected LZ (STZ, type 1 diabetic), and ZDF rats underwent carotid artery balloon injury (±10 mg PROLI/NO), and vessels were harvested at 3 and 14 d. UbcH10 was assessed by Western blotting and immunofluorescent staining. RESULTS: NO more effectively reduced UbcH10 levels in LZ versus ZDF VSMCs; however, addition of insulin and glucose dramatically potentiated the inhibitory effect of NO on UbcH10 in ZDF VSMCs. Three days after balloon injury, Western blotting showed NO decreased free UbcH10 and increased polyubiquitinated UbcH10 levels by 35% in both STZ and ZDF animals. Fourteen days after injury, immunofluorescent staining showed increased UbcH10 levels throughout the arterial wall in all animal models. NO decreased UbcH10 levels in LZ and STZ rats but not in ZDF. CONCLUSIONS: These data suggest a disconnect between UbcH10 levels and neointimal hyperplasia formation in type 2 diabetic models and contribute valuable insight regarding differential efficacy of NO in these models.


Subject(s)
Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Nitric Oxide/pharmacology , Ubiquitin-Conjugating Enzymes/blood , Animals , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Hyperplasia , Male , Muscle, Smooth, Vascular/chemistry , Neointima/pathology , Rats , Rats, Zucker , Streptozocin , Ubiquitination
19.
Ann Vasc Surg ; 29(4): 830-5, 2015.
Article in English | MEDLINE | ID: mdl-25595111

ABSTRACT

BACKGROUND: The pathogenesis of fibromuscular dysplasia (FMD) remains poorly understood. Yet, understanding this mechanism has taken on new urgency after recent evidence indicating that FMD is not as rare as previously thought. We speculated that hormonal receptors in the walls of dysplastic renal arteries were implicated in the pathogenesis of FMD. METHODS: We undertook a pilot prospective case-control study comparing histologic findings from renal arteries that were surgically removed in 2 patient groups. The case group included 6 samples from FMD patients who underwent surgery for stenosis or aneurysm caused by FMD. The control group included 3 FMD-free patients who underwent nephrectomy for nonvascular causes. Surgical specimens were sent to the histology laboratory. FMD was defined preoperatively using conventional radiologic criteria and was confirmed by histologic examination. RESULTS: Immunohistochemical staining detected intense progesterone receptor expression in the nuclei of smooth muscle cells in FMD patients. No progesterone receptor expression was found in the FMD-free patients. Estrogen receptor expression was not noted in the 2 groups. CONCLUSIONS: This preliminary finding may suggest that progesterone plays a key role in the pathogenesis of FMD and opens the fields of genetic and therapeutic approaches.


Subject(s)
Cell Nucleus/chemistry , Fibromuscular Dysplasia/metabolism , Muscle, Smooth, Vascular/chemistry , Myocytes, Smooth Muscle/chemistry , Receptors, Progesterone/analysis , Adult , Case-Control Studies , Cell Nucleus/pathology , Female , Fibromuscular Dysplasia/pathology , Fibromuscular Dysplasia/surgery , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/surgery , Myocytes, Smooth Muscle/pathology , Pilot Projects , Renal Artery/chemistry , Renal Artery/pathology
20.
J Vasc Surg ; 60(1): 202-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23911244

ABSTRACT

INTRODUCTION: Human saphenous vein (HSV) is the most widely used bypass conduit for peripheral and coronary vascular reconstructions. However, outcomes are limited by a high rate of intimal hyperplasia (IH). HSV undergoes a series of ex vivo surgical manipulations prior to implantation, including hydrostatic distension, marking, and warm ischemia in solution. We investigated the impact of surgical preparation on HSV cellular function and development of IH in organ culture. We hypothesized that oxidative stress is a mediator of HSV dysfunction. METHODS: HSV was collected from patients undergoing vascular bypass before and after surgical preparation. Smooth muscle and endothelial function were measured using a muscle bath. Endothelial preservation was assessed with immunohistochemical staining. An organ culture model was used to investigate the influence of surgical preparation injury on the development of IH. Superoxide levels were measured using a high-performance liquid chromatography-based assay. The influence of oxidative stress on HSV physiologic responses was investigated by exposing HSV to hydrogen peroxide (H2O2). RESULTS: Surgical vein graft preparation resulted in smooth muscle and endothelial dysfunction, endothelial denudation, diminished endothelial nitric oxide synthase staining, development of increased IH, and increased levels of reactive oxygen species. Experimental induction of oxidative stress in unmanipulated HSV by treatment with H2O2 promoted endothelial dysfunction. Duration of storage time in solution did not contribute to smooth muscle or endothelial dysfunction. CONCLUSIONS: Surgical vein graft preparation causes dysfunction of the smooth muscle and endothelium, endothelial denudation, reduced endothelial nitric oxide synthase expression, and promotes IH in organ culture. Moreover, increased levels of reactive oxygen species are produced and may promote further vein graft dysfunction. These results argue for less injurious means of preparing HSV prior to autologous transplantation into the arterial circulation.


Subject(s)
Endothelium, Vascular/physiopathology , Muscle, Smooth, Vascular/physiopathology , Saphenous Vein/pathology , Saphenous Vein/transplantation , Tunica Intima/pathology , Aged , Endothelium, Vascular/chemistry , Endothelium, Vascular/pathology , Female , Humans , Hydrogen Peroxide/pharmacology , Hyperplasia/pathology , Male , Middle Aged , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/pathology , Nitric Oxide Synthase/analysis , Organ Culture Techniques , Oxidative Stress/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Reactive Oxygen Species/metabolism , Saphenous Vein/metabolism , Time Factors , Vascular Surgical Procedures/methods , Warm Ischemia
SELECTION OF CITATIONS
SEARCH DETAIL