Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.383
Filter
1.
Acta Pharmacol Sin ; 45(5): 975-987, 2024 May.
Article in English | MEDLINE | ID: mdl-38279042

ABSTRACT

Endothelium-dependent contraction (EDC) exists in blood vessels of normotensive animals, but is exaggerated in hypertension. An early signal in EDC is cytosolic Ca2+ rise in endothelial cells. In this study we investigated the functional role of Orai1, a major endothelial cell Ca2+ entry channel, in EDC. Hypertension model was established in WT mice by intake of L-NNA in the drinking water (0.5 g/L) for 4 weeks or osmotic pump delivery of Ang II (1.5 mg·kg-1·d-1) for 2 weeks. In TRPC5 KO mice, the concentration of L-NNA and Ang II were increased to 1 g/L or 2 mg·kg-1·d-1, respectively. Arterial segments were prepared from carotid arteries and aortas, and EDC was elicited by acetylcholine in the presence of Nω-nitro-L-arginine methyl ester. We showed that low concentration of acetylcholine (3-30 nM) initiated relaxation in phenylephrine-precontracted carotid arteries of both normotensive and hypertensive mice, while high concentration of acetylcholine (0.1-2 µM) induced contraction. Application of selective Orai1 inhibitors AnCoA4 (100 µM) or YM58483 (400 nM) had no effect on ACh-induced relaxation but markedly reduced acetylcholine-induced EDC. We found that EDC was increased in hypertensive mice compared with that of normotensive mice, which was associated with increased Orai1 expression in endothelial cells of hypertensive mice. Compared to TRPC5 and TRPV4, which were also involved in EDC, endothelial cell Orai1 had relatively greater contribution to EDC than either TRPC5 or TRPV4 alone. We identified COX-2, followed by PGF2α, PGD2 and PGE2 as the downstream signals of Orai1/TRPC5/TRPV4. In conclusion, Orai1 coordinates together with TRPC5 and TRPV4 in endothelial cells to regulate EDC responses. This study demonstrates a novel function of Orai1 in EDC in both normotensive and hypertensive mice, thus providing a general scheme about the control of EDC by Ca2+-permeable channels.


Subject(s)
Carotid Arteries , Endothelial Cells , Endothelium, Vascular , Hypertension , Mice, Inbred C57BL , Mice, Knockout , ORAI1 Protein , TRPC Cation Channels , Animals , ORAI1 Protein/metabolism , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Carotid Arteries/drug effects , Carotid Arteries/metabolism , TRPC Cation Channels/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Acetylcholine/pharmacology , Angiotensin II/pharmacology , Vasoconstriction/drug effects , TRPV Cation Channels/metabolism
2.
Cardiovasc Diabetol ; 21(1): 19, 2022 02 05.
Article in English | MEDLINE | ID: mdl-35123483

ABSTRACT

BACKGROUND: Since sodium-glucose cotransporter 2 (SGLT2) inhibitors have a pleiotropic antiatherogenic effect, they are expected to attenuate the progression of atherosclerosis. However, whether SGLT2 inhibitors affect the tissue characteristics of the human arterial wall remains unclear. This study aimed to evaluate the effects of tofogliflozin, a selective SGLT2 inhibitor, on the tissue characteristics of the human arterial wall in type 2 diabetes (T2DM) patients without apparent cardiovascular disease (CVD). METHODS: The present study was a post hoc analysis based on data obtained from the Using Tofogliflozin for Possible Better Intervention against Atherosclerosis for Type 2 Diabetes Patients (UTOPIA) trial, which was a multicenter prospective, randomized, open-label, blinded-endpoint study conducted to evaluate the efficacy of tofogliflozin in preventing the progression of atherosclerosis in patients with T2DM. We evaluated the longitudinal change in the ultrasonic tissue characteristics of the carotid wall using gray-scale median (GSM), an established index of ultrasonic tissue characteristics. The right and left intima-medial areas were delineated, and the GSM values were evaluated (right GSM-CCA and left GSM-CCA). The average values of the right and left carotid arteries were defined as "mean GSM-CCA value." RESULTS: In a mixed-effects model for repeated measures, mean GSM-CCA, along with the right and left GSM-CCA values, did not significantly change in either the tofogliflozin (n = 168) or conventional treatment group (n = 169). In addition, the tofogliflozin and conventional treatment groups did not significantly differ regarding the change of the mean GSM-CCA (mean difference [95% CI] - 1.24[- 3.87, 1.38], P = 0.35), along with the right (mean difference [95% CI] - 2.33[- 5.70, 1.05], P = 0.18) and the left GSM-CCA (mean difference [95% CI] - 0.29 [- 3.53, 2.95], P = 0.86) values. Similar findings were obtained even after adjusting for traditional cardiovascular risk factors and/or the administration of drugs at baseline. CONCLUSIONS: The tissue characteristics of the carotid arterial wall did not change in either the tofogliflozin or conventional treatment group during the 104-week treatment period, and there was no significant difference between the treatment groups. Clinical trial registration UMIN000017607 ( https://www.umin.ac.jp/icdr/index.html ).


Subject(s)
Benzhydryl Compounds/therapeutic use , Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Adult , Aged , Benzhydryl Compounds/adverse effects , Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/diagnostic imaging , Carotid Intima-Media Thickness , Diabetes Mellitus, Type 2/diagnosis , Female , Glucosides/adverse effects , Humans , Japan , Male , Middle Aged , Plaque, Atherosclerotic , Prospective Studies , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Time Factors , Treatment Outcome
3.
Biomed Pharmacother ; 147: 112632, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35045351

ABSTRACT

BACKGROUND: In recent years, several trials investigated the role of anti-inflammatory agents in reducing cardiovascular events. Trehalose is a natural disaccharide able to reduce inflammation by enhancing macrophage autophagic activity. This action has been demonstrated to attenuate atherosclerotic plaque development in various pro-atherogenic animal models. However, at present, no data about the efficacy of this compound in human subjects have been published. METHODS: We performed a randomized, double-blind trial involving 15 patients with history of myocardial infarction and evidence of systemic inflammation (defined as C-reactive protein > 2 mg/L). The patients were randomly assigned, in 2:1 ratio, to receive either intravenous trehalose (15 g once weekly) or placebo for 12 weeks. The primary efficacy end-point was the change in arterial wall inflammation, assessed by quantifying 18F-FDG PET/CT uptake in carotid arteries and ascending aorta. RESULTS: The MDS TBR change of the index vessel at 3-month follow-up was not significant in treatment and placebo groups. Furthermore, we could not demonstrate any significant difference between the trehalose group and control group in changes of cIMT from baseline to 3 months in the overall population. No significant changes in echocardiographic measurement were noted after trehalose treatment. Except for the change in urea level in placebo group (31.00 ± 6.59 vs. 25.60 ± 6.402 P = 0.038) no other changes were detected after treatment. Also, there was a significant difference between changes in alanine aminotransferase (ALT) trehalose and placebo groups. CONCLUSION: This was the first study that specifically assessed the effects of intravenous trehalose on atherogenesis in human subjects. Trehalose treatment was characterized by an optimal safety profile, but no significant reduction in arterial wall inflammation could be observed. This might be a consequence of the small sample size of this trial. Larger studies are needed to better assess the efficacy of this compound in this clinical context.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/pathology , Myocardial Infarction/pathology , Trehalose/pharmacology , Aorta/drug effects , Carotid Arteries/drug effects , Coronary Artery Disease/pathology , Double-Blind Method , Humans , Vascular Diseases/pathology
4.
Eur J Vasc Endovasc Surg ; 63(2): 336-346, 2022 02.
Article in English | MEDLINE | ID: mdl-34916111

ABSTRACT

OBJECTIVE: Hypertension is a major risk factor for intimal hyperplasia (IH) and re-stenosis following vascular and endovascular interventions. Preclinical studies suggest that hydrogen sulphide (H2S), an endogenous gasotransmitter, limits re-stenosis. While there is no clinically available pure H2S releasing compound, the sulfhydryl containing angiotensin converting enzyme inhibitor zofenopril is a source of H2S. Here, it was hypothesised that zofenopril, due to H2S release, would be superior to other non-sulfhydryl containing angiotensin converting enzyme inhibitors (ACEi) in reducing intimal hyperplasia. METHODS: Spontaneously hypertensive male Cx40 deleted mice (Cx40-/-) or wild type (WT) littermates were randomly treated with enalapril 20 mg or zofenopril 30 mg. Discarded human vein segments and primary human smooth muscle cells (SMCs) were treated with the active compound enalaprilat or zofenoprilat. IH was evaluated in mice 28 days after focal carotid artery stenosis surgery and in human vein segments cultured for seven days ex vivo. Human primary smooth muscle cell (SMC) proliferation and migration were studied in vitro. RESULTS: Compared with control animals (intima/media thickness 2.3 ± 0.33 µm), enalapril reduced IH in Cx40-/- hypertensive mice by 30% (1.7 ± 0.35 µm; p = .037), while zofenopril abrogated IH (0.4 ± 0.16 µm; p < .002 vs. control and p > .99 vs. sham operated Cx40-/- mice). In WT normotensive mice, enalapril had no effect (0.9665 ± 0.2 µm in control vs. 1.140 ± 0.27 µm; p > .99), while zofenopril also abrogated IH (0.1623 ± 0.07 µm; p < .008 vs. control and p > .99 vs. sham operated WT mice). Zofenoprilat, but not enalaprilat, also prevented IH in human vein segments ex vivo. The effect of zofenopril on carotid and SMCs correlated with reduced SMC proliferation and migration. Zofenoprilat inhibited the mitogen activated protein kinase and mammalian target of rapamycin pathways in SMCs and human vein segments. CONCLUSION: Zofenopril provides extra beneficial effects compared with non-sulfhydryl ACEi in reducing SMC proliferation and re-stenosis, even in normotensive animals. These findings may hold broad clinical implications for patients suffering from vascular occlusive diseases and hypertension.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Captopril/analogs & derivatives , Carotid Stenosis/drug therapy , Hypertension/complications , Tunica Intima/pathology , Animals , Blood Pressure/drug effects , Captopril/administration & dosage , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Cells, Cultured , Disease Models, Animal , Humans , Hydrogen Sulfide/metabolism , Hyperplasia/drug therapy , Hyperplasia/pathology , Hypertension/drug therapy , Male , Mice , Myocytes, Smooth Muscle , Organ Culture Techniques , Primary Cell Culture , Tunica Intima/drug effects , Veins/drug effects , Veins/pathology
5.
Life Sci ; 286: 120048, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34655604

ABSTRACT

AIMS: Clinically, Cerebralcare Granule® (CG) has been widely utilized to treat various types of headache, chronic cerebral insufficiency and other diseases, and the effect is significant. Clinical studies have shown that CG can significantly relieve vascular dementia (VaD), however, the molecular mechanisms haven't been established. To clear the therapeutic mechanisms of CG against VaD, a hypothesis was proposed that CG could treat neurovascular injury by inhibiting the production of lipocalin-2 (LCN 2). MAIN METHODS: 90 dementia rats were selected by water maze test and randomly divided into 6 groups, including nimodipine (NM), CG L (low dose) (0.314 g kg-1), CG H (high dose) (0.628 g kg-1), and combined group (CG + NM). And in vitro neuronal cell OGD modeling to evaluate the effect of CG on JAK2/STAT3. KEY FINDINGS: CG could significantly shorten the escape latency of two-vessel occlusion (2-VO) rats, increase their exploratory behavior, alleviate the symptoms of VaD and improve the ultrastructural pathological damage of neurovascular unit and accelerate the recovery of cerebral blood perfusion. CG combined with NM is better than NM alone. It was further showed that CG could inhibit the pathogenicity of LCN 2 through JAK2/STAT3 pathway and suppress the production of inflammatory cytokines. It plays a role in the protection of cerebral microvasculature and BBB in 2-VO rats. SIGNIFICANCE: Taken together, there data has supported notion that CG can protect the integrity of cerebral blood vessels and BBB and improve cognitive impairment through mainly inhibiting LCN 2, which provides scientific evidence for clinical application.


Subject(s)
Cognitive Dysfunction/drug therapy , Drugs, Chinese Herbal/metabolism , Lipocalin-2/metabolism , Animals , Carotid Arteries/drug effects , China , Cognitive Dysfunction/physiopathology , Dementia, Vascular/prevention & control , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Lipocalin-2/physiology , Male , Maze Learning/drug effects , Neuroprotective Agents/pharmacology , Nimodipine/metabolism , Nimodipine/pharmacology , Rats , Rats, Sprague-Dawley
6.
Sci Rep ; 11(1): 20674, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667238

ABSTRACT

Vascular restenosis remains a major problem in patients with coronary artery disease (CAD) and peripheral artery disease (PAD). Neointimal hyperplasia, defined by post-procedure proliferation and migration of vascular smooth muscle cells (VSMCs) is a key underlying pathology. Here we investigated the role of Interleukin 11 (IL-11) in a mouse model of injury-related plaque development. Apoe-/- mice were fed a hyperlipidaemic diet and subjected to carotid wire injury of the right carotid. Mice were injected with an anti-IL11 antibody (X203), IgG control antibody or buffer. We performed ultrasound analysis to assess vessel wall thickness and blood velocity. Using histology and immunofluorescence approaches, we determined the effects of IL-11 inhibition on VSMC and macrophages phenotypes and fibrosis. Treatment of mice with carotid wire injury using X203 significantly reduced post-endothelial injury vessel wall thickness, and injury-related plaque, when compared to control. Immunofluorescence staining of the injury-related plaque showed that X203 treatment did not reduce macrophage numbers, but reduced the number of VSMCs and lowered matrix metalloproteinase 2 (MMP2) levels and collagen content in comparison to control. X203 treatment was associated with a significant increase in smooth muscle protein 22α (SM22α) positive cells in injury-related plaque compared to control, suggesting preservation of the contractile VSMC phenotype. Interestingly, X203 also reduced the collagen content of uninjured carotid arteries as compared to IgG, showing an additional effect on hyperlipidemia-induced arterial remodeling in the absence of mechanical injury. Therapeutic inhibition of IL-11 reduced vessel wall thickness, attenuated neointimal hyperplasia, and has favorable effects on vascular remodeling following wire-induced endothelial injury. This suggests IL-11 inhibition as a potential novel therapeutic approach to reduce arterial stenosis following revascularization in CAD and PAD patients.


Subject(s)
Antibodies, Neutralizing/pharmacology , Carotid Arteries/drug effects , Carotid Artery Injuries/drug therapy , Hyperplasia/drug therapy , Interleukin-11/metabolism , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Disease Models, Animal , Hyperplasia/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neointima/drug therapy , Neointima/metabolism , Vascular Remodeling/drug effects
7.
Sci Rep ; 11(1): 18094, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508162

ABSTRACT

Severe burns result in cardiovascular dysfunction, but responses in the peripheral vasculature are unclear. We hypothesize that severe burns disturb arterial contractility through acute changes in adrenergic and cholinergic receptor function. To address this, we investigated the changes in carotid artery contractility and relaxation following a severe burn. Thirty-four adult Sprague-Dawley male rats received a 40% total body surface area (TBSA) scald burn and fluid resuscitation using the Parkland formula. Control animals received sham burn procedure. Animals were serially euthanized between 6 h and 14 days after burn and endothelium-intact common carotid arteries were used for ex vivo force/relaxation measurements. At 6 h after burn, carotid arteries from burned animals demonstrated a > 50% decrease in cumulative dose-responses to norepinephrine (p < 0.05) and to 10-7 M angiotensin II (p < 0.05). Notably, pre-constricted carotid arteries also demonstrated reduced relaxation responses to acetylcholine (p < 0.05) 6 h after burn, but not to sodium nitroprusside. Histologic examination of cross-sectional planes revealed significant increases in carotid artery wall thickness in burned rats at 6 h versus 3 days, with increased collagen expression in tunica media at 3 days (p < 0.05). Carotid artery dysfunction occurs within 6 h after severe burn, demonstrating decreased sensitivity to adrenergic- and angiotensin II-induced vasoconstriction and acetylcholine-induced relaxation.


Subject(s)
Burns/physiopathology , Carotid Arteries/physiopathology , Muscle, Smooth, Vascular/physiopathology , Vasoconstriction , Animals , Biomarkers , Burns/diagnosis , Burns/etiology , Burns/metabolism , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Immunohistochemistry , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Potassium Chloride/pharmacology , Rats , Vasoconstriction/drug effects
8.
Medicine (Baltimore) ; 100(35): e26201, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34477113

ABSTRACT

ABSTRACT: This retrospective study appraised the preventive effect of statin after carotid artery stenting (CAS).Records were extracted for 100 patients with CAS surgery indicator, aged between 20 and 75 years old, and treated for statin. The cohort study included treatment group (statin and routine treatment) and control group (routine treatment), each group 50 patients. Outcomes consisted of degree of nerve defect (as measured by National Institute of Health Stroke Scale), lipid profiles (mg/dL), and CAS complications within 30 days after surgery.After treatment, there were no significant differences in National Institute of Health Stroke Scale, lipid profiles, and mortality rate between 2 groups. However, significant differences in total cholesterol (mg/dL, P = .03), low-density lipoprotein (mg/dL, P = .01), transient ischemic attack (P = .03), ischemic stroke (P = .04), and cardiac complications (P = .03) were identified within 30 days after CAS between 2 groups.The results of this study showed that prior statin treatment may be effective for the prevention of CAS complications.


Subject(s)
Carotid Arteries/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/standards , Stents/standards , Aged , Carotid Arteries/physiopathology , China/epidemiology , Cohort Studies , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Male , Middle Aged , Retrospective Studies , Risk Factors , Statistics, Nonparametric , Stents/statistics & numerical data , Treatment Outcome
9.
Acta Biochim Biophys Sin (Shanghai) ; 53(10): 1310-1320, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34409427

ABSTRACT

Endothelial dysfunction is the pathological basis of atherosclerosis. Incomplete understanding of endothelial dysfunction etiology has impeded drug development for this devastating disease despite the currently available therapies. Floralozone, an aroma flavor, specifically exists in rabbit ear grass. Recently, floralozone has been demonstrated to inhibit atherosclerosis, but the underlying mechanisms are undefined. The present study was undertaken to explore whether floralozone pharmacologically targets endothelial dysfunction and therefore exerts therapeutic effects on atherosclerosis. The Na+/H+ exchanger 1 (NHE1), a channel protein, plays a vital role in atherosclerosis. Whether NHE1 is involved in the therapeutic effects of floralozone on endothelial dysfunction has yet to be further answered. By performing oil red staining and hematoxylin-eosin staining, vascular functional study, and oxidative stress monitoring, we found that floralozone not only reduced the size of carotid atherosclerotic plaque but also prevented endothelial dysfunction in atherosclerotic rats. NHE1 expression was upregulated in the inner membrane of carotid arteries and H2O2-induced primary rat aortic endothelial cells. Inspiringly, floralozone prevented the upregulation of NHE1 in vivo and in vitro. Notably, the administration of NHE1 activator LiCl significantly weakened the protective effect of floralozone on endothelial dysfunction in vivo and in vitro. Our study demonstrated that floralozone exerted its protective effect on endothelial dysfunction in atherosclerosis by ameliorating NHE1. NHE1 maybe a drug target for the treatment of atherosclerosis, and floralozone may be an effective drug to meet the urgent needs of atherosclerosis patients by dampening NHE1.


Subject(s)
Atherosclerosis , Endothelium, Vascular , Plant Extracts , Protective Agents , Sodium-Hydrogen Exchanger 1 , Animals , Male , Aorta/cytology , Aorta/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Carotid Arteries/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/drug effects , Oxidation-Reduction/drug effects , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control , Protective Agents/pharmacology , Protective Agents/therapeutic use , Rats, Sprague-Dawley , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/metabolism
10.
Toxicol Ind Health ; 37(9): 535-546, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34396864

ABSTRACT

Formaldehyde (FA) is a ubiquitous organic preservative used in several industries and represents an occupational health hazard. Short-term exposure to FA can increase oxidative stress and cause a decrease in conduit vessel function. These decrements in vascular function may extend to the arterial architecture, predisposing individuals to increased risk of cardiovascular disease. The purpose of this study was to investigate the impact of an acute 90-minute FA exposure period (259 ± 95 ppb) on indices of arterial architecture. Arterial stiffness and carotid distensibility as determined by central pressures, augmentation index (AIx), and carotid-femoral pulse wave velocity (cfPWV) (n=13F, 24 ± 1 year) as well as carotid stiffness and intima media thickness (IMT) (n = 9F, 23 ± 1 year) were assessed prior to (Pre-FA) and immediately following (Post-FA) exposure to FA in human cadaver dissection laboratories. Central pressures and cfPWV (Pre-FA: 5.2 ± 0.8 m.s-1, Post-FA: 5.2 ± 1.1 m s-1) were unchanged by acute FA exposure (p > 0.05). Carotid stiffness parameters and distension were unchanged by acute FA exposure (p > 0.05), although distensibility (Pre-FA: 33.9 ± 10.5[10-3*kPa-1], Post-FA: 25.9 ± 5.5[10-3*kPa-1], p < 0.05), and IMT (Pre-FA: 0.42 ± 0.05 mm, Post-FA: 0.51 ± 0.11 mm, p < 0.05) decreased and increased, respectively. Individual Pre- to Post-FA changes in these markers of arterial architecture did not correlate with levels of FA exposure ([FA]: 20-473 ppb) (p > 0.05). Our group previously found vascular function decrements following acute FA exposure in human cadaver laboratories; here we found that carotid distensibility and intima media thickness are altered following FA exposure.


Subject(s)
Carotid Arteries/drug effects , Formaldehyde/adverse effects , Occupational Exposure/adverse effects , Vascular Stiffness/drug effects , Adolescent , Cadaver , Carotid Intima-Media Thickness , Female , Formaldehyde/pharmacology , Humans , Respiratory Hypersensitivity
11.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34264848

ABSTRACT

During operations, surgical mesh is commonly fixed on tissues through fasteners such as sutures and staples. Attributes of surgical mesh include biocompatibility, flexibility, strength, and permeability, but sutures and staples may cause stress concentration and tissue damage. Here, we show that the functions of surgical mesh can be significantly broadened by developing a family of materials called hydrogel-mesh composites (HMCs). The HMCs retain all the attributes of surgical mesh and add one more: adhesion to tissues. We fabricate an HMC by soaking a surgical mesh with a precursor, and upon cure, the precursor forms a polymer network of a hydrogel, in macrotopological entanglement with the fibers of the surgical mesh. In a surgery, the HMC is pressed onto a tissue, and the polymers in the hydrogel form covalent bonds with the tissue. To demonstrate the concept, we use a poly(N-isopropylacrylamide) (PNIPAAm)/chitosan hydrogel and a polyethylene terephthalate (PET) surgical mesh. In the presence a bioconjugation agent, the chitosan and the tissue form covalent bonds, and the adhesion energy reaches above 100 J⋅m-2 At body temperature, PNIPAAm becomes hydrophobic, so that the hydrogel does not swell and the adhesion is stable. Compared with sutured surgical mesh, the HMC distributes force over a large area. In vitro experiments are conducted to study the application of HMCs to wound closure, especially on tissues under high mechanical stress. The performance of HMCs on dynamic living tissues is further investigated in the surgery of a sheep.


Subject(s)
Hydrogels/pharmacology , Surgical Mesh , Wound Healing , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Female , Liver/drug effects , Liver/pathology , Sheep , Tissue Adhesives/pharmacology , Wound Healing/drug effects
12.
Am J Physiol Heart Circ Physiol ; 321(1): H185-H196, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34114892

ABSTRACT

We assessed the efficacy of oral supplementation with the flavanoid apigenin on arterial function during aging and identified critical mechanisms of action. Young (6 mo) and old (27 mo) C57BL/6N mice (model of arterial aging) consumed drinking water containing vehicle (0.2% carboxymethylcellulose; 10 young and 7 old) or apigenin (0.5 mg/mL in vehicle; 10 young and 9 old) for 6 wk. In vehicle-treated animals, isolated carotid artery endothelium-dependent dilation (EDD), bioassay of endothelial function, was impaired in old versus young (70% ± 9% vs. 92% ± 1%, P < 0.0001) due to reduced nitric oxide (NO) bioavailability. Old mice had greater arterial reactive oxygen species (ROS) production and oxidative stress (higher nitrotyrosine) associated with greater nicotinamide adenine dinucleotide phosphate oxidase (oxidant enzyme) and lower superoxide dismutase 1 and 2 (antioxidant enzymes); ex vivo administration of Tempol (antioxidant) restored EDD to young levels, indicating ROS-mediated suppression of EDD. Old animals also had greater aortic stiffness as indicated by higher aortic pulse wave velocity (PWV, 434 ± 9 vs. 346 ± 5 cm/s, P < 0.0001) due to greater intrinsic aortic wall stiffness associated with lower elastin levels and higher collagen, advanced glycation end products (AGEs), and proinflammatory cytokine abundance. In old mice, apigenin restored EDD (96% ± 2%) by increasing NO bioavailability, normalized arterial ROS, oxidative stress, and antioxidant expression, and abolished ROS inhibition of EDD. Moreover, apigenin prevented foam cell formation in vitro (initiating step in atherosclerosis) and mitigated age-associated aortic stiffening (PWV 373 ± 5 cm/s) by normalizing aortic intrinsic wall stiffness, collagen, elastin, AGEs, and inflammation. Thus, apigenin is a promising therapeutic for arterial aging.NEW & NOTEWORTHY Our study provides novel evidence that oral apigenin supplementation can reverse two clinically important indicators of arterial dysfunction with age, namely, vascular endothelial dysfunction and large elastic artery stiffening, and prevents foam cell formation in an established cell culture model of early atherosclerosis. Importantly, our results provide extensive insight into the biological mechanisms of apigenin action, including increased nitric oxide bioavailability, normalization of age-related increases in arterial ROS production and oxidative stress, reversal of age-associated aortic intrinsic mechanical wall stiffening and adverse remodeling of the extracellular matrix, and suppression of vascular inflammation. Given that apigenin is commercially available as a dietary supplement in humans, these preclinical findings provide the experimental basis for future translational studies assessing the potential of apigenin to treat arterial dysfunction and reduce cardiovascular disease risk with aging.


Subject(s)
Aging/metabolism , Endothelium, Vascular/drug effects , Inflammation/metabolism , Oxidative Stress/drug effects , Spirostans/pharmacology , Vascular Stiffness/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Endothelium, Vascular/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
13.
Biosci Rep ; 41(6)2021 05 28.
Article in English | MEDLINE | ID: mdl-33969376

ABSTRACT

It has been demonstrated that trimethylamine N-oxide (TMAO) serves as a driver of atherosclerosis, suggesting that reduction of TMAO level might be a potent method to prevent the progression of atherosclerosis. Herein, we explored the role of TMAO in the stability of carotid atherosclerotic plaques and disclosed the underlying mechanisms. The unstable carotid artery plaque models were established in C57/BL6 mice. L-carnitine (LCA) and methimazole (MMI) administration were applied to increase and reduce TMAO levels. Hematoxylin and eosin (H&E) staining, Sirius red, Perl's staining, Masson trichrome staining and immunohistochemical staining with CD68 staining were used for histopathology analysis of the carotid artery plaque. M1 and M2 macrophagocyte markers were assessed by RT-PCR to determine the polarization of RAW264.7 cells. MMI administration for 2 weeks significantly decreased the plaque area, increased the thickness of the fibrous cap and reduced the size of the necrotic lipid cores, whereas 5-week of administration of MMI induced intraplate hemorrhage. LCA treatment further deteriorated the carotid atherosclerotic plaque but with no significant difference. In mechanism, we found that TMAO treatment impaired the M2 polarization and efferocytosis of RAW264.7 cells with no obvious effect on the M1 polarization. In conclusion, the present study demonstrated that TMAO reduction enhanced the stability of carotid atherosclerotic plaque through promoting macrophage M2 polarization and efferocytosis.


Subject(s)
Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Enzyme Inhibitors/pharmacology , Macrophages/drug effects , Methimazole/pharmacology , Methylamines/metabolism , Phagocytosis/drug effects , Plaque, Atherosclerotic , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Disease Models, Animal , Down-Regulation , Fibrosis , Humans , Jurkat Cells , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Oxygenases/antagonists & inhibitors , Oxygenases/metabolism , Phenotype , RAW 264.7 Cells
14.
Eur J Pharmacol ; 904: 174155, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33971178

ABSTRACT

The methylglyoxal elicits diverse adverse effects on the body. Uridine diphosphate, an extracellular nucleotide, plays an important role as a signaling molecule controlling vascular tone. This study aimed to evaluate the relationship between methylglyoxal and uridine diphosphate-induced carotid arterial contraction in rats. Additionally, we examined whether p38 mitogen-activated protein kinase (MAPK) would involve such responses. Organ baths were conducted to determine vascular reactivity in isolated carotid arterial rings, and western blotting was used for protein analysis. Treatment with methylglyoxal to carotid arterial rings showed concentration-dependent augmentation to uridine diphosphate-induced contraction in the absence and presence of NG-nitro-L-arginine, which is a nitric oxide synthase inhibitor, whereas, methylglyoxal did not affect serotonin- or isotonic high K+-induced contraction in the presence of a nitric oxide synthase inhibitor. Under nitric oxide synthase inhibition, SB203580, which is a selective p38 MAPK inhibitor, suppressed uridine diphosphate-induced contraction in both the control and methylglyoxal-treated groups, and the difference in uridine diphosphate-induced contraction was abolished by SB203580 treatment. The levels of phosphorylated p38 MAPK were increased by methylglyoxal in carotid arteries, not only under the basal condition but also under uridine diphosphate stimulation. The suppression of uridine diphosphate-induced contraction by a highly selective cell-permeable protein kinase C inhibitor bisindolylmaleimide I was observed in the methylglyoxal-treated group but not in the controls. Moreover, methylglyoxal-induced augmentation of uridine diphosphate-induced contraction was prevented by N-acetyl-L-cysteine. These results suggest that methylglyoxal could enhance uridine diphosphate-induced contraction in rat carotid arteries and may be caused by activation of p38 MAPK and protein kinase C and increased oxidative stress.


Subject(s)
Carotid Arteries/drug effects , Carotid Arteries/metabolism , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Pyruvaldehyde/pharmacology , Uridine Diphosphate/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Free Radical Scavengers/pharmacology , Imidazoles/pharmacology , Male , Muscle, Skeletal/blood supply , Oxidative Stress/drug effects , Potassium/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats, Wistar , Serotonin/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Am J Obstet Gynecol ; 225(5): 544.e1-544.e9, 2021 11.
Article in English | MEDLINE | ID: mdl-33887239

ABSTRACT

BACKGROUND: Nifedipine is a widely used drug in pregnancies complicated by maternal hypertensive disorders that can be associated with placental insufficiency and fetal hypoxemia. The evidence regarding fetal myocardial responses to nifedipine in hypoxemia is limited. OBJECTIVE: We hypothesized that nifedipine would not impair fetal sheep cardiac function under hypoxemic environment. In particular, we investigated the effects of nifedipine on fetal ventricular functional parameters and cardiac output. STUDY DESIGN: A total of 21 chronically instrumented fetal sheep at 122 to 134 gestational days (term, 145 days) were included in this study. Fetal cardiac function was evaluated by measuring global longitudinal strain, indices describing ventricular systolic and diastolic function, and cardiac outputs using two-dimensional speckle tracking and tissue and spectral pulsed-wave Doppler echocardiography. Fetal carotid artery blood pressure and blood gas values were invasively monitored. After baseline data collection, fetal hypoxemia was induced by maternal hyperoxygenation. After hypoxemia phase data collection, 9 fetuses received nifedipine infusion, and 12 fetuses received saline infusion. Data were collected 30 and 120 minutes after the infusion was started. After 120 minutes of data collection, maternal and fetal oxygenation were normalized, and normoxemia phase data were collected, while infusion was continued. RESULTS: Hypoxemia decreased fetal carotid artery mean arterial pressure from 40 (8) mm Hg to 35 (8) mm Hg (P<.007), and left ventricular global longitudinal strain showed less deformation than at baseline (P=.001). Under hypoxemia, nifedipine caused a reduction in right ventricular global longitudinal strain (P<.05), a decrease in right ventricular isovolumic relaxation velocity and its deceleration (P<.01) indicating diastolic dysfunction, and a drop in right ventricular cardiac output (P<.05). Nifedipine did not alter fetal left ventricular functional parameters or cardiac output. When normoxemia was restored, fetal right ventricular functional parameters and cardiac output returned to baseline level. CONCLUSION: In hypoxemic fetus, nifedipine impaired right ventricular function and reduced its cardiac output. The detrimental effects of nifedipine on fetal right ventricular function were abolished, when normoxemia was restored. Our findings suggest that in a hypoxemic environment nifedipine triggers detrimental effects on fetal right ventricular function.


Subject(s)
Calcium Channel Blockers/adverse effects , Cardiac Output/drug effects , Fetal Hypoxia/complications , Nifedipine/adverse effects , Ventricular Dysfunction, Right/chemically induced , Animals , Arterial Pressure/drug effects , Carotid Arteries/drug effects , Diastole/drug effects , Echocardiography, Doppler, Pulsed , Fetal Monitoring , Models, Animal , Sheep
16.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33906946

ABSTRACT

Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.


Subject(s)
Blood-Brain Barrier/drug effects , Immunity, Innate/genetics , Inflammation/genetics , Mannitol/pharmacology , Animals , Blood-Brain Barrier/metabolism , Carotid Arteries/drug effects , Cell Adhesion/drug effects , Cell Adhesion Molecules/blood , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/genetics , Chemokines/blood , Cytokines/blood , Endothelial Cells/drug effects , Humans , Inflammation/blood , Rats , Tight Junctions/drug effects , Tight Junctions/genetics
17.
Medicine (Baltimore) ; 100(10): e25114, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33725908

ABSTRACT

ABSTRACT: It remains uncertain whether statin/ezetimibe combination therapy serves as a useful and equivalent alternative to statin monotherapy for reducing atherosclerotic plaque inflammation. The aim of the present study was to compare the effects of statin/ezetimibe combination therapy and statin monotherapy on carotid atherosclerotic plaque inflammation using 18F-fluorodeoxyglucose (18FDG) positron emission tomography (PET)/computed tomography (CT) imaging. Data were pooled from 2 clinical trials that used serial 18FDG PET/CT examination to investigate the effects of cholesterol-lowering therapy on carotid atherosclerotic plaque inflammation. The primary outcome was the percent change in the target-to-background ratio (TBR) of the index vessel in the most diseased segment (MDS) at 6-month follow-up. Baseline characteristics were largely similar between the 2 groups. At the 6-month follow-up, the MDS TBR of the index vessel significantly decreased in both groups. The percent change in the MDS TBR of the index vessel (primary outcome) did not differ significantly between the 2 groups (-8.41 ±â€Š15.9% vs -8.08 ±â€Š17.0%, respectively, P = .936). Likewise, the percent change in the whole vessel TBR of the index vessel did not differ significantly between the 2 groups. There were significant decreases in total and LDL cholesterol levels in both groups at follow-up (P < .001). There were no significant correlations between the percent changes in MDS TBR of the index vessel, changes in the lipid, and high-sensitive C-reactive protein levels. The reduction in carotid atherosclerotic plaque inflammation by statin/ezetimibe combination therapy was equivalent to that by the statin monotherapy.


Subject(s)
Acute Coronary Syndrome/drug therapy , Carotid Artery Diseases/drug therapy , Ezetimibe, Simvastatin Drug Combination/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Plaque, Atherosclerotic/drug therapy , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/immunology , Aged , C-Reactive Protein/analysis , Carotid Arteries/diagnostic imaging , Carotid Arteries/drug effects , Carotid Arteries/immunology , Carotid Artery Diseases/blood , Carotid Artery Diseases/complications , Carotid Artery Diseases/immunology , Cholesterol, LDL/blood , Clinical Trials as Topic , Datasets as Topic , Female , Follow-Up Studies , Humans , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Male , Middle Aged , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/immunology , Rosuvastatin Calcium/administration & dosage , Simvastatin/administration & dosage
18.
Biochem Biophys Res Commun ; 548: 127-133, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33640605

ABSTRACT

Diabetes is a major risk factor for the development of cardiovascular disease. Diabetic patients have a higher incidence of restenosis following endovascular therapy than non-diabetic patients. Melatonin is primarily synthesized and secreted by the pineal gland and plays an important protective anti-inflammatory and antioxidant role in a variety of cardiovascular diseases. However, no studies to date have evaluated the underlying effects and molecular mechanisms of melatonin on diabetes-related restenosis. Herein, we used an in vivo model of diabetes-related restenosis and an in vitro model of high glucose-cultured vascular smooth muscle cells to investigate the anti-restenosis effect and signaling mechanisms induced by melatonin treatment. The present study provides the first evidence that melatonin attenuates restenosis following vascular injury in diabetic rats. We further investigated the underlying molecular mechanisms both in vivo and in vitro. The data suggest that the Nrf2 signaling pathway is an important molecular target for melatonin-mediated inhibition of diabetes-related restenosis after vascular injury. These findings indicate that melatonin may represent a potential candidate for the prevention or treatment of vascular diseases and restenosis following endovascular therapy, especially in diabetic patients.


Subject(s)
Coronary Restenosis/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Melatonin/therapeutic use , NF-E2-Related Factor 2/metabolism , Signal Transduction , Vascular System Injuries/drug therapy , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coronary Restenosis/complications , Coronary Restenosis/pathology , Cytoprotection/drug effects , Diabetes Mellitus, Experimental/complications , Glucose/toxicity , Heme Oxygenase-1/metabolism , Hyperplasia , Male , Melatonin/pharmacology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vascular System Injuries/complications , Vascular System Injuries/pathology
19.
Sci Rep ; 11(1): 4126, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602971

ABSTRACT

Individuals with chronic kidney disease are at an increased risk for cardiovascular disease. This risk may partially be explained by a chronic inflammatory state in these patients, reflected by increased arterial wall and cellular inflammation. Statin treatment decreases cardiovascular risk and arterial inflammation in non-CKD subjects. In patients with declining kidney function, cardiovascular benefit resulting from statin therapy is attenuated, possibly due to persisting inflammation. In the current study, we assessed the effect of statin treatment on arterial wall and cellular inflammation. Fourteen patients with chronic kidney disease stage 3 or 4, defined by an estimated Glomerular Filtration Rate between 15 and 60 mL/min/1.73 m2, without cardiovascular disease were included in a single center, open label study to assess the effect of atorvastatin 40 mg once daily for 12 weeks (NTR6896). At baseline and at 12 weeks of treatment, we assessed arterial wall inflammation by 18F-fluoro-deoxyglucose positron-emission tomography computed tomography (18F-FDG PET/CT) and the phenotype of circulating monocytes were assessed. Treatment with atorvastatin resulted in a 46% reduction in LDL-cholesterol, but this was not accompanied by an attenuation in arterial wall inflammation in the aorta or carotid arteries, nor with changes in chemokine receptor expression of circulating monocytes. Statin treatment does not abolish arterial wall or cellular inflammation in subjects with mild to moderate chronic kidney disease. These results imply that CKD-associated inflammatory activity is mediated by factors beyond LDL-cholesterol and specific anti-inflammatory interventions might be necessary to further dampen the inflammatory driven CV risk in these subjects.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atorvastatin/therapeutic use , Cardiovascular Diseases/drug therapy , Inflammation/drug therapy , Renal Insufficiency, Chronic/diet therapy , Aged , Aorta/drug effects , Aorta/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Cholesterol, LDL/metabolism , Female , Fluorodeoxyglucose F18/administration & dosage , Heart Disease Risk Factors , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/metabolism , Male , Middle Aged , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals/administration & dosage , Renal Insufficiency, Chronic/metabolism , Risk Factors
20.
J Stroke Cerebrovasc Dis ; 30(3): 105580, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33387888

ABSTRACT

OBJECTIVES: Inflammation plays an important role in the development of atherosclerotic vascular disease, which is the leading cause of morbidity and mortality in the adult population. Several clinical trials have shown that suppression of the inflammatory response can delay or decrease the atherosclerotic process. The aim of this study was to investigate carotid intima-media thickness (CIMT) between patients with chronic disease history plus gout using colchicine and patients with cardiovascular risk factors. MATERIALS AND METHODS: In total, 102 patients (85 female, 17 male) were included. There were two groups in the study: Group 1 - patients with chronic diseases including cardiovascular risk factors plus gout using colchicine (0,5 mg twice a day); and Group 2 - patients with chronic diseases including cardiovascular risk factors only. All patients underwent ultrasonography for the measurement of CIMT. Additionally, the serum concentrations of C-reactive protein (CRP) and the levels of lipids such as cholesterol, triglyceride, LDL, HDL were measured. RESULTS: The mean age of patients was 62.35±6.68 years and 64.27±5.32 years in Group 1 and Group 2, respectively. There was also no statistically significant difference in the levels of lipids between groups (p>0.05). The value of CIMT and CRP in Group 1 and Group 2 were 0.98±0.20 and 0.26±0.14, 1.18±0.15 and 0.58±0.42, respectively. There was a statistically significant difference between groups (p<0.05). The colchicine group was found to have a statistically significant lowering of CIMT and CRP compared to the non-colchicine group. CONCLUSIONS: It appears that colchicine in addition to statins and other standard treatments is an effective treatment for the interception of cardiovascular and cerebrovascular events in patients with cardiovascular risk factors.


Subject(s)
Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Carotid Intima-Media Thickness , Colchicine/therapeutic use , Gout Suppressants/therapeutic use , Gout/drug therapy , Aged , Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/complications , Carotid Artery Diseases/diagnostic imaging , Cross-Sectional Studies , Female , Gout/complications , Gout/diagnosis , Heart Disease Risk Factors , Humans , Lipids/blood , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Risk Assessment , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...