Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.439
Filter
1.
J Am Heart Assoc ; 13(10): e033998, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38726925

ABSTRACT

BACKGROUND: The vasoconstrictor effects of angiotensin II via type 1 angiotensin II receptors in vascular smooth muscle cells are well established, but the direct effects of angiotensin II on vascular endothelial cells (VECs) in vivo and the mechanisms how VECs may mitigate angiotensin II-mediated vasoconstriction are not fully understood. The present study aimed to explore the molecular mechanisms and pathophysiological relevance of the direct actions of angiotensin II on VECs in kidney and brain microvessels in vivo. METHODS AND RESULTS: Changes in VEC intracellular calcium ([Ca2+]i) and nitric oxide (NO) production were visualized by intravital multiphoton microscopy of cadherin 5-Salsa6f mice or the endothelial uptake of NO-sensitive dye 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate, respectively. Kidney fibrosis by unilateral ureteral obstruction and Ready-to-use adeno-associated virus expressing Mouse Renin 1 gene (Ren1-AAV) hypertension were used as disease models. Acute systemic angiotensin II injections triggered >4-fold increases in VEC [Ca2+]i in brain and kidney resistance arterioles and capillaries that were blocked by pretreatment with the type 1 angiotensin II receptor inhibitor losartan, but not by the type 2 angiotensin II receptor inhibitor PD123319. VEC responded to acute angiotensin II by increased NO production as indicated by >1.5-fold increase in 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate fluorescence intensity. In mice with kidney fibrosis or hypertension, the angiotensin II-induced VEC [Ca2+]i and NO responses were significantly reduced, which was associated with more robust vasoconstrictions, VEC shedding, and microthrombi formation. CONCLUSIONS: The present study directly visualized angiotensin II-induced increases in VEC [Ca2+]i and NO production that serve to counterbalance agonist-induced vasoconstriction and maintain residual organ blood flow. These direct and endothelium-specific angiotensin II effects were blunted in disease conditions and linked to endothelial dysfunction and the development of vascular pathologies.


Subject(s)
Angiotensin II , Brain , Calcium , Hypertension , Kidney , Microvessels , Nitric Oxide , Vasoconstriction , Animals , Nitric Oxide/metabolism , Angiotensin II/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/drug therapy , Kidney/blood supply , Kidney/metabolism , Calcium/metabolism , Vasoconstriction/drug effects , Microvessels/metabolism , Microvessels/drug effects , Microvessels/pathology , Brain/metabolism , Brain/blood supply , Mice , Disease Models, Animal , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice, Inbred C57BL , Calcium Signaling/drug effects
2.
Respir Res ; 25(1): 205, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730297

ABSTRACT

BACKGROUND: Obesity is the main risk factor leading to the development of various respiratory diseases, such as asthma and pulmonary hypertension. Pulmonary microvascular endothelial cells (PMVECs) play a significant role in the development of lung diseases. Aconitate decarboxylase 1 (Acod1) mediates the production of itaconate, and Acod1/itaconate axis has been reported to play a protective role in multiple diseases. However, the roles of Acod1/itaconate axis in the PMVECs of obese mice are still unclear. METHODS: mRNA-seq was performed to identify the differentially expressed genes (DEGs) between high-fat diet (HFD)-induced PMVECs and chow-fed PMVECs in mice (|log2 fold change| ≥ 1, p ≤ 0.05). Free fatty acid (FFA) was used to induce cell injury, inflammation and mitochondrial oxidative stress in mouse PMVECs after transfection with the Acod1 overexpressed plasmid or 4-Octyl Itaconate (4-OI) administration. In addition, we investigated whether the nuclear factor erythroid 2-like 2 (Nrf2) pathway was involved in the effects of Acod1/itaconate in FFA-induced PMVECs. RESULTS: Down-regulated Acod1 was identified in HFD mouse PMVECs by mRNA-seq. Acod1 expression was also reduced in FFA-treated PMVECs. Acod1 overexpression inhibited cell injury, inflammation and mitochondrial oxidative stress induced by FFA in mouse PMVECs. 4-OI administration showed the consistent results in FFA-treated mouse PMVECs. Moreover, silencing Nrf2 reversed the effects of Acod1 overexpression and 4-OI administration in FFA-treated PMVECs, indicating that Nrf2 activation was required for the protective effects of Acod1/itaconate. CONCLUSION: Our results demonstrated that Acod1/Itaconate axis might protect mouse PMVECs from FFA-induced injury, inflammation and mitochondrial oxidative stress via activating Nrf2 pathway. It was meaningful for the treatment of obesity-caused pulmonary microvascular endotheliopathy.


Subject(s)
Carboxy-Lyases , Endothelial Cells , Lung , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Obesity , Succinates , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Carboxy-Lyases/metabolism , Carboxy-Lyases/genetics , Obesity/metabolism , Obesity/complications , Male , Succinates/pharmacology , Lung/metabolism , Lung/drug effects , Lung/pathology , Lung/blood supply , Cells, Cultured , Microvessels/metabolism , Microvessels/drug effects , Microvessels/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hydro-Lyases
3.
Exp Gerontol ; 192: 112452, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38718888

ABSTRACT

Ischemic stroke rapidly increases the expression level of vascular endothelial growth factor (VEGF), which promotes neovascularization during hypoxia. However, the effect and mechanism of VEGF intervention on cerebrovascular formation remain unclear. Therefore, our research discussed the protective effect of exogenous VEGF on cells in hypoxia environment in cerebral microvascular endothelial cells, simulating ischemic stroke in hypoxic environment. Firstly, we detected the proliferation and apoptosis of cerebral microvascular endothelial cells under hypoxia environment, as well the expression levels of VEGF-E, vascular endothelial growth factor re-ceptor-2 (VEGFR-2), BCL2, PRKCE and PINK1. Moreover, immunofluorescence and western blotting were used to verify the regulation of exogenous VEGF-E on VEGFR-2 expression in hypoxic or normal oxygen environment. Lastly, we manipulated the concentration of VEGF-E in the culture medium to investigate its impact on phospholipase Cγ1 (PLCγ1)/extracellular signaling regulatory protein kinase (ERK) -1/2 and protein kinase B (AKT) pathways. Additionally, we employed a PLCγ1 inhibitor (U73122) to investigate its impact on proliferation and PLCγ1/ERK pathways. The results show that hypoxia inhibited the proliferation of cerebral microvascular endothelial cells, promoted cell apoptosis, significantly up-regulated the expression of VEGF-E, VEGFR-2, PRKCE and PINK1, but down-regulated the expression of BCL2. Interference from exogenous VEGF-E activated PLCγ1/ERK-1/2 and AKT pathways, promoting cell proliferation and inhibiting apoptosis of hypoxic brain microvascular endothelial cells. In summary, exogenous VEGF-E prevents hypoxia-induced damage to cerebral microvascular endothelial cells by activating the PLCγ1/ERK and AKT pathways. This action inhibits the apoptosis pathway in hypoxic cerebral microvascular endothelial cells, thereby safeguarding the blood-brain barrier and the nervous system.


Subject(s)
Apoptosis , Cell Hypoxia , Endothelial Cells , Phospholipase C gamma , Proto-Oncogene Proteins c-akt , Signal Transduction , Phospholipase C gamma/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Brain/blood supply , Vascular Endothelial Growth Factor Receptor-2/metabolism , Rats , Microvessels/drug effects , Cells, Cultured , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
4.
Photodiagnosis Photodyn Ther ; 46: 104070, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38599306

ABSTRACT

Standardized measurements are needed to obtain reliable data, therefore the aim of this letter is to clarify some points concerning chronic central serous chorioretinopathy.


Subject(s)
Central Serous Chorioretinopathy , Photochemotherapy , Central Serous Chorioretinopathy/drug therapy , Humans , Photochemotherapy/methods , Chronic Disease , Photosensitizing Agents/therapeutic use , Tomography, Optical Coherence , Microvessels/drug effects
5.
J Bioenerg Biomembr ; 56(3): 193-204, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38446318

ABSTRACT

Blood-brain barrier breakdown and ROS overproduction are important events during the progression of ischemic stroke aggravating brain damage. Geraniol, a natural monoterpenoid, possesses anti-apoptotic, cytoprotective, anti-oxidant, and anti-inflammatory activities. Our study aimed to investigate the effect and underlying mechanisms of geraniol in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced human brain microvascular endothelial cells (HBMECs). Apoptosis, caspase-3 activity, and cytotoxicity of HBMECs were evaluated using TUNEL, caspase-3 activity, and CCK-8 assays, respectively. The permeability of HBMECs was examined using FITC-dextran assay. Reactive oxygen species (ROS) production was measured using the fluorescent probe DCFH-DA. The protein levels of zonula occludens-1 (ZO-1), occludin, claudin-5, ß-catenin, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) were determined by western blotting. Geraniol showed no cytotoxicity in HBMECs. Geraniol and ROS scavenger N-acetylcysteine (NAC) both attenuated OGD/R-induced apoptosis and increase of caspase-3 activity and the permeability to FITC-dextran in HBMECs. Geraniol relieved OGD/R-induced ROS accumulation and decrease of expression of ZO-1, occludin, claudin-5, and ß-catenin in HBMECs. Furthermore, we found that geraniol activated Nrf2/HO-1 pathway to inhibit ROS in HBMECs. In conclusion, geraniol attenuated OGD/R-induced ROS-dependent apoptosis and permeability in HBMECs through activating the Nrf2/HO-1 pathway.


Subject(s)
Acyclic Monoterpenes , Apoptosis , Endothelial Cells , Glucose , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Reactive Oxygen Species , Humans , Apoptosis/drug effects , Acyclic Monoterpenes/pharmacology , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Glucose/metabolism , Heme Oxygenase-1/metabolism , Oxygen/metabolism , Brain/metabolism , Brain/blood supply , Microvessels/metabolism , Microvessels/pathology , Microvessels/drug effects
6.
J Cereb Blood Flow Metab ; 44(5): 680-688, 2024 May.
Article in English | MEDLINE | ID: mdl-38420777

ABSTRACT

The accumulation of the microtubule-associated tau protein in and around blood vessels contributes to brain microvascular dysfunction through mechanisms that are incompletely understood. Delivery of nutrients to active neurons in the brain relies on capillary calcium (Ca2+) signals to direct blood flow. The initiation and amplification of endothelial cell Ca2+ signals require an intact microtubule cytoskeleton. Since tau accumulation in endothelial cells disrupts native microtubule stability, we reasoned that tau-induced microtubule destabilization would impair endothelial Ca2+ signaling. We tested the hypothesis that tau disrupts the regulation of local cerebral blood flow by reducing endothelial cell Ca2+ signals and endothelial-dependent vasodilation. We used a pathogenic soluble tau peptide (T-peptide) model of tau aggregation and mice with genetically encoded endothelial Ca2+ sensors to measure cerebrovascular endothelial responses to tau exposure. T-peptide significantly attenuated endothelial Ca2+ activity and cortical capillary blood flow in vivo. Further, T-peptide application constricted pressurized cerebral arteries and inhibited endothelium-dependent vasodilation. This study demonstrates that pathogenic tau alters cerebrovascular function through direct attenuation of endothelial Ca2+ signaling and endothelium-dependent vasodilation.


Subject(s)
Calcium Signaling , Cerebrovascular Circulation , Microvessels , Vasodilation , tau Proteins , Animals , Vasodilation/drug effects , Calcium Signaling/drug effects , Mice , tau Proteins/metabolism , Microvessels/metabolism , Microvessels/drug effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Brain/blood supply , Brain/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Calcium/metabolism
7.
Arterioscler Thromb Vasc Biol ; 43(8): e291-e302, 2023 08.
Article in English | MEDLINE | ID: mdl-37317849

ABSTRACT

The ECM (extracellular matrix) is a 3-dimensional network that supports cellular responses and maintains structural tissue integrity in healthy and pathological conditions. The interactions between ECM and cells trigger signaling cascades that lead to phenotypic changes and structural and compositional turnover of the ECM, which in turn regulates vascular cell behavior. Hydrogel biomaterials are a powerful platform for basic and translational studies and clinical applications due to their high swelling capacity and exceptional versatility in compositions and properties. This review highlights recent developments and uses of engineered natural hydrogel platforms that mimic the ECM and present defined biochemical and mechanical cues for vascularization. Specifically, we focus on modulating vascular cell stimulation and cell-ECM/cell-cell interactions in the microvasculature that are the established biomimetic microenvironment.


Subject(s)
Biomimetic Materials , Extracellular Matrix , Hydrogels , Microvessels , Neovascularization, Physiologic , Tissue Engineering , Hydrogels/chemistry , Hydrogels/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Microvessels/drug effects , Microvessels/physiology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Humans , Tissue Engineering/methods , Tissue Scaffolds , Gene Transfer Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Animals
8.
Clin Hemorheol Microcirc ; 83(3): 287-292, 2023.
Article in English | MEDLINE | ID: mdl-36591655

ABSTRACT

Cannabinoids play critical roles in human pathophysiology through the cannabinoid (CB) receptors and non-CB receptors on variety of cells, tissues, and organs. Microvasculature with the inside bloodstream containing the plasmatic and cellular components exerts multiple functions in maintaining tissue and organ physiology through microcirculation. This review focusses on the impact of cannabinoids on the microvasculature, including mechanisms mediated by both CB receptor-related pathways and CB receptor-independent pathways.


Subject(s)
Cannabinoids , Microcirculation , Microvessels , Receptors, Cannabinoid , Humans , Cannabinoids/pharmacology , Microvessels/drug effects , Receptor, Cannabinoid, CB2 , Microcirculation/drug effects
9.
Front Endocrinol (Lausanne) ; 13: 973058, 2022.
Article in English | MEDLINE | ID: mdl-36060954

ABSTRACT

Protein kinase C (PKC) is a family of serine/threonine protein kinases, the activation of which plays an important role in the development of diabetic microvascular complications. The activation of PKC under high-glucose conditions stimulates redox reactions and leads to an accumulation of redox stress. As a result, various types of cells in the microvasculature are influenced, leading to changes in blood flow, microvascular permeability, extracellular matrix accumulation, basement thickening and angiogenesis. Structural and functional disorders further exacerbate diabetic microvascular complications. Here, we review the roles of PKC in the development of diabetic microvascular complications, presenting evidence from experiments and clinical trials.


Subject(s)
Diabetic Angiopathies , Protein Kinase C , Capillary Permeability/drug effects , Capillary Permeability/physiology , Diabetes Mellitus , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Humans , Microvessels/drug effects , Microvessels/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Kinase C/adverse effects , Protein Kinase C/metabolism
10.
N Engl J Med ; 387(12): 1075-1088, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36129997

ABSTRACT

BACKGROUND: Data are lacking on the comparative effectiveness of commonly used glucose-lowering medications, when added to metformin, with respect to microvascular and cardiovascular disease outcomes in persons with type 2 diabetes. METHODS: We assessed the comparative effectiveness of four commonly used glucose-lowering medications, added to metformin, in achieving and maintaining a glycated hemoglobin level of less than 7.0% in participants with type 2 diabetes. The randomly assigned therapies were insulin glargine U-100 (hereafter, glargine), glimepiride, liraglutide, and sitagliptin. Prespecified secondary outcomes with respect to microvascular and cardiovascular disease included hypertension and dyslipidemia, confirmed moderately or severely increased albuminuria or an estimated glomerular filtration rate of less than 60 ml per minute per 1.73 m2 of body-surface area, diabetic peripheral neuropathy assessed with the Michigan Neuropathy Screening Instrument, cardiovascular events (major adverse cardiovascular events [MACE], hospitalization for heart failure, or an aggregate outcome of any cardiovascular event), and death. Hazard ratios are presented with 95% confidence limits that are not adjusted for multiple comparisons. RESULTS: During a mean 5.0 years of follow-up in 5047 participants, there were no material differences among the interventions with respect to the development of hypertension or dyslipidemia or with respect to microvascular outcomes; the mean overall rate (i.e., events per 100 participant-years) of moderately increased albuminuria levels was 2.6, of severely increased albuminuria levels 1.1, of renal impairment 2.9, and of diabetic peripheral neuropathy 16.7. The treatment groups did not differ with respect to MACE (overall rate, 1.0), hospitalization for heart failure (0.4), death from cardiovascular causes (0.3), or all deaths (0.6). There were small differences with respect to rates of any cardiovascular disease, with 1.9, 1.9, 1.4, and 2.0 in the glargine, glimepiride, liraglutide, and sitagliptin groups, respectively. When one treatment was compared with the combined results of the other three treatments, the hazard ratios for any cardiovascular disease were 1.1 (95% confidence interval [CI], 0.9 to 1.3) in the glargine group, 1.1 (95% CI, 0.9 to 1.4) in the glimepiride group, 0.7 (95% CI, 0.6 to 0.9) in the liraglutide group, and 1.2 (95% CI, 1.0 to 1.5) in the sitagliptin group. CONCLUSIONS: In participants with type 2 diabetes, the incidences of microvascular complications and death were not materially different among the four treatment groups. The findings indicated possible differences among the groups in the incidence of any cardiovascular disease. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; GRADE ClinicalTrials.gov number, NCT01794143.).


Subject(s)
Cardiovascular Diseases , Diabetes Complications , Diabetes Mellitus, Type 2 , Glycated Hemoglobin , Hypoglycemic Agents , Metformin , Albuminuria/etiology , Albuminuria/prevention & control , Blood Glucose/analysis , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Comparative Effectiveness Research , Diabetes Complications/etiology , Diabetes Complications/prevention & control , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/diagnosis , Diabetic Neuropathies/etiology , Diabetic Neuropathies/prevention & control , Drug Therapy, Combination , Dyslipidemias/etiology , Dyslipidemias/prevention & control , Glomerular Filtration Rate , Glycated Hemoglobin/analysis , Heart Failure/etiology , Heart Failure/prevention & control , Humans , Hypertension/etiology , Hypertension/prevention & control , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Insulin Glargine/adverse effects , Insulin Glargine/therapeutic use , Liraglutide/adverse effects , Liraglutide/therapeutic use , Metformin/adverse effects , Metformin/therapeutic use , Microvessels/drug effects , Sitagliptin Phosphate/adverse effects , Sitagliptin Phosphate/therapeutic use , Sulfonylurea Compounds/adverse effects , Sulfonylurea Compounds/therapeutic use
11.
Biomed Pharmacother ; 151: 113165, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35609370

ABSTRACT

OBJECTIVE: To investigate the efficacy of a paeoniflorin-sodium alginate (SA)-gelatin skin scaffold for treating diabetic wound in a rat model. METHODS: Bioinks were prepared using various percentages of paeoniflorin in the total weight of a solution containing SA and gelatin. Skin scaffolds containing 0%, 1%, 3%, 5%, and 10% paeoniflorin were printed using 3D bioprinting technology, and scaffold microstructure was observed with scanning electron microscopy. Skin scaffolds were then used in rats with diabetic wounds. H&E staining, Masson staining, and immunohistochemical staining for IL-1ß and CD31 were performed on days 7 and 14. RESULTS: All skin scaffolds had a mesh-like structure with uniform pore distribution. Wounds healed well in each group, with the 1% and 3% groups demonstrating the most complete healing. H&E staining showed that skin accessory organs had appeared in each group. On day 7, collagen deposition in the 3% group was higher than in the other groups (P<0.05), and IL-1ß infiltration was lower in the 10% group than in the 3% group (P = 0.002). On day 14, IL-1ß infiltration was not significantly different between the 10% and 3% groups (P = 0.078). The CD31 level was higher in the 3% group than in the other groups on days 7 and 14 (P<0.05). CONCLUSION: A 3% paeoniflorin-SA-gelatin skin scaffold promoted the healing of diabetic wounds in rats. This scaffold promoted collagen deposition and microvascular regeneration and demonstrated anti-inflammatory properties, suggesting that this scaffold type could be used to treat diabetic wounds.


Subject(s)
Alginates , Diabetes Complications , Gelatin , Glucosides , Skin , Tissue Scaffolds , Alginates/administration & dosage , Alginates/therapeutic use , Animals , Collagen/metabolism , Diabetes Complications/complications , Diabetes Complications/therapy , Diabetes Mellitus , Disease Models, Animal , Gelatin/administration & dosage , Gelatin/therapeutic use , Glucosides/administration & dosage , Glucosides/therapeutic use , Microvessels/drug effects , Microvessels/physiology , Monoterpenes/administration & dosage , Monoterpenes/therapeutic use , Printing, Three-Dimensional , Rats , Skin/blood supply , Skin/drug effects , Skin/injuries , Wound Healing/drug effects , Wound Healing/physiology , Wounds and Injuries/complications , Wounds and Injuries/physiopathology , Wounds and Injuries/therapy
12.
Arch Biochem Biophys ; 720: 109173, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35300940

ABSTRACT

The effect of Esculetin on pyroptosis and its possible mechanism in endothelium were explored. 10 µg/mL LPS and 0.5 mM ATP were used to stimulate the rat intestinal microvascular endothelial cells. Then add different concentrations of Esculetin (20µM, 40 µM) to the culture medium containing LPS and ATP culturing for 24 h. The expression of p-NF-κB p65, NF-κB p65, I-κB, p-I-κB, NLRP3, ASC, caspase-1, and gasdermin-D were detected by Western blot, and the release level of IL-18 and IL-1ß were measured by ELISA. The NLRP3 inhibitor MCC950 was used at the concentration of 10 µM for 4 h to disentangle the potential mechanism of the influence of Esculetin on pyroptosis. In our experiments, the expression of gasdermin-d and important proteins of NF-κB and NLRP3 signaling pathways were inhibited by Esculetin. Besides, Esculetin also attenuated the morphological changes like swelling rupture and pores on the membrane caused by pyroptosis thereby protecting cells from being damaged by pyroptosis. Combining with the effect of Esculetin on proteins above and its protective effect on cell morphology, we believe that Esculetin has an anti-pyroptosis effect. The inhibiting pyroptosis effects mentioned above are similar to MCC950, which means the anti-pyroptosis effects of Esculetin are associated with the NLRP3 signaling pathway. In conclusion, Esculetin inhibits the pyroptosis of microvascular endothelial cells through the NF-κB/NLFP3 signaling pathway and is expected to be conducive in treating pyroptosis-related diseases.


Subject(s)
Endothelial Cells , Microvessels , NF-kappa B , Pyroptosis , Umbelliferones , Adenosine Triphosphate , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipopolysaccharides/pharmacology , Microvessels/cytology , Microvessels/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Rats , Signal Transduction , Umbelliferones/pharmacology
13.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055038

ABSTRACT

Obesity is a worrisomely escalating public health problem globally and one of the leading causes of morbidity and mortality from noncommunicable disease. The epidemiological link between obesity and a broad spectrum of cardiometabolic disorders has been well documented; however, the underlying pathophysiological mechanisms are only partially understood, and effective treatment options remain scarce. Given its critical role in glucose metabolism, skeletal muscle has increasingly become a focus of attention in understanding the mechanisms of impaired insulin function in obesity and the associated metabolic sequelae. We examined the current evidence on the relationship between microvascular dysfunction and insulin resistance in obesity. A growing body of evidence suggest an intimate and reciprocal relationship between skeletal muscle microvascular and glucometabolic physiology. The obesity phenotype is characterized by structural and functional changes in the skeletal muscle microcirculation which contribute to insulin dysfunction and disturbed glucose homeostasis. Several interconnected etiologic molecular mechanisms have been suggested, including endothelial dysfunction by several factors, extracellular matrix remodelling, and induction of oxidative stress and the immunoinflammatory phenotype. We further correlated currently available pharmacological agents that have deductive therapeutic relevance to the explored pathophysiological mechanisms, highlighting a potential clinical perspective in obesity treatment.


Subject(s)
Insulin Resistance , Microcirculation , Microvessels/physiopathology , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Biomarkers , Disease Management , Disease Susceptibility , Endothelium, Vascular/metabolism , Energy Metabolism , Extracellular Matrix , Gene Expression Regulation , Homeostasis , Humans , Insulin/metabolism , Microvessels/drug effects , Microvessels/metabolism , Molecular Targeted Therapy , Obesity/therapy , Oxidation-Reduction , Vascular Diseases/etiology , Vascular Diseases/metabolism
14.
Am J Physiol Cell Physiol ; 322(3): C338-C353, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35044858

ABSTRACT

The small conductance calcium-activated potassium channel (KCa2.3) has long been recognized for its role in mediating vasorelaxation through the endothelium-derived hyperpolarization (EDH) response. Histone deacetylases (HDACs) have been implicated as potential modulators of blood pressure and histone deacetylase inhibitors (HDACi) are being explored as therapeutics for hypertension. Herein, we show that HDACi increase KCa2.3 expression when heterologously expressed in HEK cells and endogenously expressed in primary cultures of human umbilical vein endothelial cells (HUVECs) and human intestinal microvascular endothelial cells (HIMECs). When primary endothelial cells were exposed to HDACi, KCa2.3 transcripts, subunits, and functional current are increased. Quantitative RT-PCR (qPCR) demonstrated increased KCa2.3 mRNA following HDACi, confirming transcriptional regulation of KCa2.3 by HDACs. By using pharmacological agents selective for different classes of HDACs, we discriminated between cytoplasmic and epigenetic modulation of KCa2.3. Biochemical analysis revealed an association between the cytoplasmic HDAC6 and KCa2.3 in immunoprecipitation studies. Specifically inhibiting HDAC6 increases expression of KCa2.3. In addition to increasing the expression of KCa2.3, we show that nonspecific inhibition of HDACs causes an increase in the expression of the molecular chaperone Hsp70 in endothelial cells. When Hsp70 is inhibited in the presence of HDACi, the magnitude of the increase in KCa2.3 expression is diminished. Finally, we show a slower rate of endocytosis of KCa2.3 as a result of exposure of primary endothelial cells to HDACi. These data provide the first demonstrated approach to increase KCa2.3 channel number in endothelial cells and may partially account for the mechanism by which HDACi induce vasorelaxation.


Subject(s)
Endothelial Cells/drug effects , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Intestines/blood supply , Microvessels/drug effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Endocytosis , Endothelial Cells/enzymology , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylase 6/metabolism , Humans , Membrane Potentials , Microvessels/enzymology , Small-Conductance Calcium-Activated Potassium Channels/genetics , Up-Regulation , Vasodilation
15.
Am J Physiol Renal Physiol ; 322(2): F197-F207, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35001664

ABSTRACT

KV7 channels, the voltage-gated K+ channels encoded by KCNQ genes, mediate heterogeneous vascular responses in rodents. Postnatal changes in the functional expression of KV7 channels have been reported in rodent saphenous arteries, but their physiological function in the neonatal renal vascular bed is unclear. Here, we report that, unlike adult pigs, only KCNQ1 (KV7.1) out of the five members of KCNQ genes was detected in neonatal pig renal microvessels. KCNQ1 is present in fetal pig kidneys as early as day 50 of gestation, and the level of expression remains the same up to postnatal day 21. Activation of renal vascular smooth muscle cell (SMC) KV7.1 stimulated whole cell currents, inhibited by HMR1556 (HMR), a selective KV7.1 blocker. HMR did not change the steady-state diameter of isolated renal microvessels. Similarly, intrarenal artery infusion of HMR did not alter mean arterial pressure, renal blood flow, and renal vascular resistance in the pigs. An ∼20 mmHg reduction in mean arterial pressure evoked effective autoregulation of renal blood flow, which HMR inhibited. We conclude that 1) the expression of KCNQ isoforms in porcine renal microvessels is dependent on kidney maturation, 2) KV7.1 is functionally expressed in neonatal pig renal vascular SMCs, 3) a decrease in arterial pressure up to 20 mmHg induces renal autoregulation in neonatal pigs, and 4) SMC KV7.1 does not control basal renal vascular tone but contributes to neonatal renal autoregulation triggered by a step decrease in arterial pressure.NEW & NOTEWORTHY KV7.1 is present in fetal pig kidneys as early as day 50 of gestation, and the level of expression remains the same up to postnatal day 21. KV7.1 is functionally expressed in neonatal pig renal vascular smooth muscle cells (SMCs). A decrease in arterial pressure up to 20 mmHg induces renal autoregulation in neonatal pigs. Although SMC KV7.1 does not control basal renal vascular resistance, its inhibition blunts neonatal renal autoregulation engendered by a step decrease in arterial pressure.


Subject(s)
Arterial Pressure/drug effects , Chromans/pharmacology , KCNQ1 Potassium Channel/antagonists & inhibitors , Kidney/blood supply , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Potassium Channel Blockers/pharmacology , Renal Circulation/drug effects , Sulfonamides/pharmacology , Vasodilation/drug effects , Animals , Animals, Newborn , Gene Expression Regulation, Developmental , Gestational Age , Homeostasis , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Microvessels/drug effects , Microvessels/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Sus scrofa
16.
Am J Physiol Heart Circ Physiol ; 322(2): H328-H335, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34995168

ABSTRACT

Many individuals in industrialized societies consume a high-salt, Western diet(WD); however, the effects of this diet on microcirculatory properties and glycocalyx barrier function are unknown. Young genetically heterogeneous male and female mice underwent 12 wk of normal chow (NC) diet, NC diet with 4% salt (NC4%), Western diet (WD), or WD with 4% salt (WD4%). Microcirculatory properties and glycocalyx barrier function were evaluated in the mesenteric microcirculation, using an intravital microscope equipped with an automated capture and analysis system. Total microvascular density summed across 4- to 25-µm microvessel segment diameters was lower in NC4% than in NC and WD (P < 0.05). Perfused boundary region (PBR), a marker of glycocalyx barrier function, averaged across 4- to 25-µm microvessel segment diameters was similar between NC and NC4%, as well as between WD and WD4% (P > 0.05). PBR was lower in WD and WD4% than in NC and NC4% (P < 0.05), indicating augmented glycocalyx barrier function in WD and WD4%. There were strong, inverse relationships between PBR and adiposity and blood glucose (r = -0.44 to -0.61, P < 0.05). In summary, NC4% induces deleterious effects on microvascular density, whereas WD augments glycocalyx barrier function. Interestingly, the combination of high-salt, Western diet in WD4% resulted in lower total microvascular density like NC4% and augmented glycocalyx barrier function like WD. These data suggest distinct microcirculatory adaptations to high-salt and Western diets that coincide when these diets are combined in young genetically heterogeneous male and female mice.NEW & NOTEWORTHY Many individuals in industrialized societies consume a combination of high-salt and Western diet; however, the effects of this diet on microcirculatory and glycocalyx properties are unknown. This study reveals that a high-salt diet lowers microcirculatory and glycocalyx properties, whereas a Western diet augments glycocalyx barrier function and thickness. Taken together, these data indicate that there are distinct microcirculatory adaptations to high-salt and Western diets that coincide when high-salt and Western diets are combined.


Subject(s)
Diet, Western , Glycocalyx/metabolism , Microcirculation , Sodium Chloride, Dietary/adverse effects , Adiposity , Animals , Animals, Outbred Strains , Blood Glucose/metabolism , Female , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice , Microvessels/drug effects , Microvessels/metabolism , Microvessels/physiology
17.
J Ethnopharmacol ; 283: 114705, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34655669

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Dan-Deng-Tong-Nao Capsules (DDTNC) is a Chinese patent medicine and has been used in treating cerebral ischemic stroke (IS) for a long time in China, protection of brain microvascular endothelial cells (BMECs) is the main treatment strategy. But the holistic chemical information and potential bioactive components of DDTNC on protecting BMECs and its underlying mechanism is still unclear. AIM OF THE STUDY: To identify the active ingredients of DDTNC and to explore the protective effects of DDTNC on BMECs associated with Wnt/ß-catenin pathway. MATERIALS AND METHODS: The components of DDTNC and cerebrospinal fluid containing composition of DDTNC (DDTNC-CSF) were detected by High performance liquid chromatography combined with Diode array detector (HPLC-DAD) and Ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS), respectively. The experiment rat model was established with middle cerebral artery occlusion (MCAO), the therapeutic effects of DDTNC were assessed by Longa assay and TTC staining. The cerebral micro vessel density was determined by immunofluorescence staining. The injured BMECs caused by oxygen-glucose deprivation and reperfusion (OGD/R) was used to evaluate the protective effect of cerebrospinal fluid containing composition of DDTNC (DDTNC-CSF). The cell survival rate was detected by the method of CCK-8, the intracellular Ca2+ and reactive oxygen species (ROS) was estimated by Fluo-3. Moreover, the proteins of Bax, Bcl-2, Wnt, ß-catenin, GSK-3ß was determined by Western blotting. RESULTS: The RSD values of all methodological studies were less than 3.0%. A total of 20 compounds were detected under the optimized HPLC-DAD chromatographic condition. In the UPLC-Q-TOF-MS negative mode, peak 1 and peak 2 were detecteted in DDTNC-CSF and was identified as Danshensu and Puerarin, respectively. In the UPLC-Q-TOF-MS positive mode, peak 1 and peak 3 were detecteted in DDTNC-CSF and was identified as Danshensu and Scutellarin, respectively. DDTNC significantly decreased the Longa values and infarct volume and significantly increased the cerebral microvessel density of the MCAO rats. The accumulation of intracellular Ca2+ and ROS in BMECs injured by OGD/R decreased significantly in DDTNC-CSF group. The expression of Bcl-2, ß-catenin, wnt-1 was upregulated by DDTNC-CSF and the level of Bax and GSK3ß could be downregulated by DDTNC-CSF. CONCLUSION: The present study provided a scientific basis for revealing the mechanism of DDTNC in the treatment of IS and DDTNC is expected to be an effective drug for the treatment of IS.


Subject(s)
Brain Ischemia/prevention & control , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/drug effects , Ischemic Stroke/prevention & control , Animals , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Endothelial Cells/pathology , Glucose/metabolism , Infarction, Middle Cerebral Artery , Male , Microvessels/drug effects , Microvessels/pathology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Wnt Signaling Pathway/drug effects
18.
Cardiovasc Res ; 118(1): 53-64, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33620071

ABSTRACT

It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggests that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. This review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signalling, vascular inflammation and senescence will be discussed.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Coronary Vessels/drug effects , Genetic Therapy , Heart Failure, Diastolic/therapy , Heart Failure, Systolic/therapy , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Vaccines/therapeutic use , Angiogenesis Inducing Agents/adverse effects , Animals , Coronary Circulation/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Genetic Therapy/adverse effects , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/metabolism , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/genetics , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Microcirculation/drug effects , Microvessels/metabolism , Microvessels/physiopathology , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Recovery of Function , Vaccines/adverse effects , Ventricular Function, Left/drug effects
19.
Am J Physiol Cell Physiol ; 322(2): C185-C196, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34878922

ABSTRACT

The Notch pathway regulates complex patterning events in many species and is critical for the proper formation and function of the vasculature. Despite this importance, how the various components of the Notch pathway work in concert is still not well understood. For example, NOTCH1 stabilizes homotypic endothelial junctions, but the role of NOTCH1 in heterotypic interactions is not entirely clear. NOTCH3, on the other hand, is essential for heterotypic interactions of pericytes with the endothelium, but how NOTCH3 signaling in pericytes impacts the endothelium remains elusive. Here, we use in vitro vascular models to investigate whether pericyte-induced stabilization of the vasculature requires the cooperation of NOTCH1 and NOTCH3. We observe that both pericyte NOTCH3 and endothelial NOTCH1 are required for the stabilization of the endothelium. Loss of either NOTCH3 or NOTCH1 decreases the accumulation of VE-cadherin at endothelial adherens junctions and increases the frequency of wider, more motile junctions. We found that DLL4 was the key ligand for simulating NOTCH1 activation in endothelial cells and observed that DLL4 expression in pericytes is dependent on NOTCH3. Altogether, these data suggest that an interplay between pericyte NOTCH3 and endothelial NOTCH1 is critical for pericyte-induced vascular stabilization.


Subject(s)
Endothelial Cells/metabolism , Microvessels/metabolism , Pericytes/metabolism , Receptor, Notch1/metabolism , Receptor, Notch3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , HEK293 Cells , Humans , Microvessels/cytology , Microvessels/drug effects , Pericytes/drug effects , Receptor, Notch1/agonists , Receptor, Notch3/agonists
20.
Neuroimmunomodulation ; 29(4): 349-358, 2022.
Article in English | MEDLINE | ID: mdl-34937041

ABSTRACT

INTRODUCTION: Invasion of viruses into the brain causes viral encephalitis, which can be fatal and causes permanent brain damage. The blood-brain barrier (BBB) protects the brain by excluding harmful substances and microbes. Brain microvascular endothelial cells are important components of the BBB; however, the mechanisms of antiviral reactions in these cells have not been fully elucidated. Zinc-finger antiviral protein (ZAP) is a molecule that restricts the infection of various viruses, and there are 2 major isoforms: ZAPL and ZAPS. Toll-like receptor 3 (TLR3), a pattern-recognition receptor against viral double-stranded RNA, is implicated in antiviral innate immune reactions. The aim of this study was to investigate the expression of ZAP in cultured hCMEC/D3 human brain microvascular endothelial cells treated with an authentic TLR3 agonist polyinosinic-polycytidylic acid (poly IC). METHODS: hCMEC/D3 cells were cultured and treated with poly IC. Expression of ZAPL and ZAPS mRNA was investigated using quantitative reverse transcription-polymerase chain reaction, and protein expression of these molecules was examined using western blotting. The role of nuclear factor-κB (NF-κB) was examined using the NF-κB inhibitor, SN50. The roles of interferon (IFN)-ß, IFN regulatory factor 3 (IRF3), tripartite motif protein 25 (TRIM25), and retinoic acid-inducible gene-I (RIG-I) in poly IC-induced ZAPS expression were examined using RNA interference. Propagation of Japanese encephalitis virus (JEV) was examined using a focus-forming assay. RESULTS: ZAPS mRNA and protein expression was upregulated by poly IC, whereas the change of ZAPL mRNA and protein levels was minimal. Knockdown of IRF3 or TRIM25 decreased the poly IC-induced upregulation of ZAPS, whereas knockdown of IFN-ß or RIG-I did not affect ZAPS upregulation. SN50 did not affect ZAPS expression. Knockdown of ZAP enhanced JEV propagation. CONCLUSION: ZAPL and ZAPS were expressed in hCMEC/D3 cells, and ZAPS expression was upregulated by poly IC. IRF3 and TRIM25 are involved in poly IC-induced upregulation of ZAPS. ZAP may contribute to antiviral reactions in brain microvascular endothelial cells and protect the brain from invading viruses such as JEV.


Subject(s)
Antiviral Agents , Cerebrum , Encephalitis Virus, Japanese , Endothelial Cells , Microvessels , Toll-Like Receptor 3 , Humans , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , NF-kappa B/metabolism , Poly I-C/pharmacology , RNA, Messenger/metabolism , Toll-Like Receptor 3/immunology , Zinc , Microvessels/drug effects , Microvessels/immunology , Cerebrum/blood supply , Cerebrum/immunology , Encephalitis Virus, Japanese/drug effects , Encephalitis Virus, Japanese/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...