Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.022
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 173(1): 74-89.e20, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29570999

RESUMO

A decline in capillary density and blood flow with age is a major cause of mortality and morbidity. Understanding why this occurs is key to future gains in human health. NAD precursors reverse aspects of aging, in part, by activating sirtuin deacylases (SIRT1-SIRT7) that mediate the benefits of exercise and dietary restriction (DR). We show that SIRT1 in endothelial cells is a key mediator of pro-angiogenic signals secreted from myocytes. Treatment of mice with the NAD+ booster nicotinamide mononucleotide (NMN) improves blood flow and increases endurance in elderly mice by promoting SIRT1-dependent increases in capillary density, an effect augmented by exercise or increasing the levels of hydrogen sulfide (H2S), a DR mimetic and regulator of endothelial NAD+ levels. These findings have implications for improving blood flow to organs and tissues, increasing human performance, and reestablishing a virtuous cycle of mobility in the elderly.


Assuntos
Envelhecimento , Sulfeto de Hidrogênio/metabolismo , NAD/metabolismo , Animais , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos Knockout , Microvasos/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Condicionamento Físico Animal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Sirtuína 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Eur J Immunol ; 54(8): e2350792, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38727188

RESUMO

Loss of perfusion in the burn wound might cause wound deepening and impaired healing. We previously showed persistent microvascular thrombosis coinciding with intraluminal neutrophils extracellular traps in human burned skin. This study investigates the presence of intraluminal citrullinated histone 3 (H3cit) from different cellular origins (neutrophils, monocytes, and lymphocytes) in relation to microvascular thrombosis of burn wounds. Eschar was obtained from burn patients (n = 18) 6-40 days postburn with a mean total burned body surface area of 23%. Microvascular presence of tissue factor (TF), factor XII (FXII) and thrombi was assessed by immunohistochemistry. Intramicrovascular cell death was analyzed via immunofluorescent microscopy, combining antibodies for neutrophils (MPO), monocytes (CD14), and lymphocytes (CD45) with endothelial cell markers CD31 and H3cit. Significantly increased microvascular expression of TF, FXII, and thrombi (CD31+) was found in all eschar samples compared with control uninjured skin. Release of H3cit from different cellular origins was observed in the lumen of the dermal microvasculature in the eschar tissue 7-40 days postburn, with release from neutrophilic origin being 2.7 times more abundant. Intraluminal presence of extracellular H3cit colocalizing with either MPO, CD14, or CD45 is correlated to increased microvascular thrombosis in eschar of burn patients.


Assuntos
Queimaduras , Citrulinação , Histonas , Neutrófilos , Trombose , Humanos , Queimaduras/imunologia , Queimaduras/metabolismo , Queimaduras/complicações , Histonas/metabolismo , Histonas/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Trombose/metabolismo , Trombose/imunologia , Trombose/patologia , Tromboplastina/metabolismo , Idoso , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator XII/metabolismo , Microvasos/patologia , Microvasos/imunologia , Microvasos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Pele/patologia , Pele/imunologia , Pele/metabolismo , Pele/irrigação sanguínea , Linfócitos/imunologia , Linfócitos/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/metabolismo , Adulto Jovem
3.
Am J Pathol ; 194(6): 1078-1089, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38417697

RESUMO

Ferroptosis is a new form of cell death characterized by iron-dependent lipid peroxidation. Whether ferroptosis is involved in retinal microvascular dysfunction under diabetic condition is not known. Herein, the expression of ferroptosis-related genes in patients with proliferative diabetic retinopathy and in diabetic mice was determined with quantitative RT-PCR. Reactive oxygen species, iron content, lipid peroxidation products, and ferroptosis-associated proteins in the cultured human retinal microvascular endothelial cells (HRMECs) and in the retina of diabetic mice were examined. The association of ferroptosis with the functions of endothelial cells in vitro was evaluated. After administration of ferroptosis-specific inhibitor, Fer-1, the retinal microvasculature in diabetic mice was assessed. Characteristic changes of ferroptosis-associated markers, including glutathione peroxidase 4, ferritin heavy chain 1, long-chain acyl-CoA synthetase 4, transferrin receptor protein 1, and cyclooxygenase-2, were detected in the retinal fibrovascular membrane of patients with proliferative diabetic retinopathy, cultured HRMECs, and the retina of diabetic mice. Elevated levels of reactive oxygen species, lipid peroxidation, and iron content were found in the retina of diabetic mice and in cultured HRMECs. Ferroptosis was found to be associated with HRMEC dysfunction under high-glucose condition. Inhibition of ferroptosis with specific inhibitor Fer-1 in diabetic mice significantly reduced the severity of retinal microvasculopathy. Ferroptosis contributes to microvascular dysfunction in diabetic retinopathy, and inhibition of ferroptosis might be a promising strategy for the therapy of early-stage diabetic retinopathy.


Assuntos
Retinopatia Diabética , Ferroptose , Espécies Reativas de Oxigênio , Retinopatia Diabética/patologia , Retinopatia Diabética/metabolismo , Animais , Humanos , Camundongos , Masculino , Espécies Reativas de Oxigênio/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Peroxidação de Lipídeos , Camundongos Endogâmicos C57BL , Microvasos/patologia , Microvasos/metabolismo , Ferro/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia
4.
Am J Pathol ; 194(7): 1171-1184, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38548268

RESUMO

Interactions between endothelial cells (ECs) and mural pericytes (PCs) are critical in maintaining the stability and function of the microvascular wall. Abnormal interactions between these two cell types are a hallmark of progressive fibrotic diseases such as systemic sclerosis (also known as scleroderma). However, the role of PCs in signaling microvascular dysfunction remains underexplored. We hypothesized that integrin-matrix interactions contribute to PC migration from the vascular wall and conversion into interstitial myofibroblasts. Herein, pro-inflammatory tumor necrosis factor α (TNFα) or a fibrotic growth factor [transforming growth factor ß1 (TGF-ß1)] were used to evaluate human PC inflammatory and fibrotic phenotypes by assessing their migration, matrix deposition, integrin expression, and subsequent effects on endothelial dysfunction. Both TNFα and TGF-ß1 treatment altered integrin expression and matrix protein deposition, but only fibrotic TGF-ß1 drove PC migration in an integrin-dependent manner. In addition, integrin-dependent PC migration was correlated to changes in EC angiopoietin-2 levels, a marker of vascular instability. Finally, there was evidence of changes in vascular stability corresponding to disease state in human systemic sclerosis skin. This work shows that TNFα and TGF-ß1 induce changes in PC integrin expression and matrix deposition that facilitate migration and reduce vascular stability, providing evidence that microvascular destabilization can be an early indicator of tissue fibrosis.


Assuntos
Movimento Celular , Fibrose , Integrinas , Pericitos , Escleroderma Sistêmico , Fator de Crescimento Transformador beta1 , Pericitos/metabolismo , Pericitos/patologia , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/metabolismo , Integrinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Microvasos/patologia , Microvasos/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Pele/patologia , Pele/metabolismo , Pele/irrigação sanguínea
5.
FASEB J ; 38(16): e70005, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39171967

RESUMO

Endothelial dysfunction, prevalent in cardiovascular diseases (CVDs) and linked to conditions like diabetes, hypertension, obesity, renal failure, or hypercholesterolemia, is characterized by diminished nitric oxide (NO) bioavailability-a key signaling molecule for vascular homeostasis. Current two-dimensional (2D) in vitro studies on NO synthesis by endothelial cells (ECs) lack the crucial laminar shear stress, a vital factor in modulating the NO-generating enzyme, endothelial nitric oxide synthase (eNOS), under physiological conditions. Here we developed a tracer-based metabolomics approach to measure NO-specific metabolites with mass spectrometry (MS) and show the impact of fluid flow on metabolic parameters associated with NO synthesis using 2D and 3D platforms. Specifically, we tracked the conversion of stable-isotope labeled NO substrate L-Arginine to L-Citrulline and L-Ornithine to determine eNOS activity. We demonstrated clear responses in human coronary artery endothelial cells (HCAECs) cultured with 13C6, 15N4-L-Arginine, and treated with eNOS stimulator, eNOS inhibitor, and arginase inhibitor. Analysis of downstream metabolites, 13C6, 15N3 L-Citrulline and 13C5, 15N2 L-Ornithine, revealed distinct outcomes. Additionally, we evaluated the NO metabolic status in static 2D culture and 3D microvessel models with bidirectional and unidirectional fluid flow. Our 3D model exhibited significant effects, particularly in microvessels exposed to the eNOS stimulator, as indicated by the 13C6, 15N3 L-Citrulline/13C5, 15N2 L-Ornithine ratio, compared to the 2D culture. The obtained results indicate that the 2D static culture mimics an endothelial dysfunction status, while the 3D model with a unidirectional fluid flow provides a more representative physiological environment that provides a better model to study endothelial dysfunction.


Assuntos
Células Endoteliais , Metabolômica , Microvasos , Óxido Nítrico Sintase Tipo III , Óxido Nítrico , Humanos , Óxido Nítrico/metabolismo , Metabolômica/métodos , Microvasos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Células Endoteliais/metabolismo , Arginina/metabolismo , Dispositivos Lab-On-A-Chip , Células Cultivadas , Citrulina/metabolismo
6.
J Mol Cell Cardiol ; 192: 26-35, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38734061

RESUMO

Coronary microvascular disease (CMD) and impaired coronary blood flow control are defects that occur early in the pathogenesis of heart failure in cardiometabolic conditions, prior to the onset of atherosclerosis. In fact, recent studies have shown that CMD is an independent predictor of cardiac morbidity and mortality in patients with obesity and metabolic disease. CMD is comprised of functional, structural, and mechanical impairments that synergize and ultimately reduce coronary blood flow in metabolic disease and in other co-morbid conditions, including transplant, autoimmune disorders, chemotherapy-induced cardiotoxicity, and remote injury-induced CMD. This review summarizes the contemporary state-of-the-field related to CMD in metabolic and these other co-morbid conditions based on mechanistic data derived mostly from preclinical small- and large-animal models in light of available clinical evidence and given the limitations of studying these mechanisms in humans. In addition, we also discuss gaps in current understanding, emerging areas of interest, and opportunities for future investigations in this field.


Assuntos
Comorbidade , Doenças Metabólicas , Humanos , Animais , Doenças Metabólicas/complicações , Doenças Metabólicas/epidemiologia , Circulação Coronária , Microvasos/patologia , Microvasos/metabolismo
7.
J Cell Mol Med ; 28(20): e70152, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39434201

RESUMO

The apelin receptor (APJ) is a key player in tumour angiogenesis, but its role in hepatocellular carcinoma (HCC) remains unclear. This study aims to elucidate the function of the apelin/APJ pathway in HCC using a multi-omics approach and identify potential therapeutic biomarkers. Differentially expressed genes related to the apelin/APJ axis were identified from bulk transcriptomics to reveal HCC-associated disparities. Single-cell and spatial transcriptomics were used to localize and analyse the function of these genes. Machine learning models were constructed to predict outcomes based on apelin/APJ expression, and experimental validation was conducted to explore the pathway's impact on HCC angiogenesis. Single cell analysis revealed an overexpression of APJ/Aplin in the endothelium. The stemness of endothelial cell (EC) with high apelin/APJ was enhanced, as well as the expression of TGFb, oxidative stresses and PI3K/AKT pathway genes. Spatial transcriptomics confirmed that EC populations with high APJ scores were enriched within the tumour. Machine learning models showed high prognostic accuracy. High APJ expression was linked to worse outcomes (p = 0.001), and AUC values were high (1 year, 3 year, 5 year) (0.95, 0.97, 0.98). Immune suppression and non-responsiveness of immune therapy were also seen in high-risk groups. The experimental validation showed that silencing apelin reduced angiogenesis (p < 0.05), endothelial proliferation, decreased expression of ANG2, KLF2, VEGFA and lower ERK1/2 phosphorylation. Apelin may serve as a potential therapeutic target in HCC, given its role in promoting tumour angiogenesis and poor patient outcomes.


Assuntos
Receptores de Apelina , Apelina , Carcinoma Hepatocelular , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas , Neovascularização Patológica , Transcriptoma , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/irrigação sanguínea , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , Humanos , Receptores de Apelina/metabolismo , Receptores de Apelina/genética , Apelina/genética , Apelina/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Análise de Célula Única , Transdução de Sinais , Microvasos/patologia , Microvasos/metabolismo , Perfilação da Expressão Gênica , Progressão da Doença , Prognóstico , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Masculino
8.
J Cell Mol Med ; 28(14): e18558, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39048917

RESUMO

Myocardial ischemia-reperfusion injury (MIRI) represents a critical pathology in acute myocardial infarction (AMI), which is characterized by high mortality and morbidity. Cardiac microvascular dysfunction contributes to MIRI, potentially culminating in heart failure (HF). Pigment epithelium-derived factor (PEDF), which belongs to the non-inhibitory serpin family, exhibits several physiological effects, including anti-angiogenesis, anti-inflammatory and antioxidant properties. Our study aims to explore the impact of PEDF and its functional peptide 34-mer on both cardiac microvascular perfusion in MIRI rats and human cardiac microvascular endothelial cells (HCMECs) injury under hypoxia reoxygenation (HR). It has been shown that MIRI is accompanied by ferroptosis in HCMECs. Furthermore, we investigated the effect of PEDF and its 34-mer, particularly regarding the Nrf2/HO-1 signalling pathway. Our results demonstrated that PEDF 34-mer significantly ameliorated cardiac microvascular dysfunction following MIRI. Additionally, they exhibited a notable suppression of ferroptosis in HCMECs, and these effects were mediated through activation of Nrf2/HO-1 signalling. These findings highlight the therapeutic potential of PEDF and 34-mer in alleviating microvascular dysfunction and MIRI. By enhancing cardiac microvascular perfusion and mitigating endothelial ferroptosis, PEDF and its derivative peptide represent promising candidates for the treatment of AMI.


Assuntos
Células Endoteliais , Proteínas do Olho , Ferroptose , Traumatismo por Reperfusão Miocárdica , Fator 2 Relacionado a NF-E2 , Fatores de Crescimento Neural , Serpinas , Transdução de Sinais , Serpinas/farmacologia , Serpinas/metabolismo , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Ferroptose/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Proteínas do Olho/metabolismo , Proteínas do Olho/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ratos , Heme Oxigenase-1/metabolismo , Masculino , Ratos Sprague-Dawley , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Peptídeos/farmacologia
9.
Angiogenesis ; 27(3): 461-474, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38780883

RESUMO

The presence of atherosclerotic plaque vessels is a critical factor in plaque destabilization. This may be attributable to the leaky phenotype of these microvessels, although direct proof for this notion is lacking. In this study, we investigated molecular and cellular patterns of stable and hemorrhaged human plaque to identify novel drivers of intraplaque vessel dysfunction. From transcriptome data of a human atherosclerotic lesion cohort, we reconstructed a co-expression network, identifying a gene module strongly and selectively correlated with both plaque microvascular density and inflammation. Spectrin Beta Non-Erythrocytic 1 (sptbn1) was identified as one of the central hubs of this module (along with zeb1 and dock1) and was selected for further study based on its predominant endothelial expression. Silencing of sptbn1 enhanced leukocyte transmigration and vascular permeability in vitro, characterized by an increased number of focal adhesions and reduced junctional VE-cadherin. In vivo, sptbn1 knockdown in zebrafish impaired the development of the caudal vein plexus. Mechanistically, increased substrate stiffness was associated with sptbn1 downregulation in endothelial cells in vitro and in human vessels. Plaque SPTBN1 mRNA and protein expression were found to correlate with an enhanced presence of intraplaque hemorrhage and future cardiovascular disease (CVD) events during follow-up. In conclusion, we identify SPTBN1 as a central hub gene in a gene program correlating with plaque vascularisation. SPTBN1 was regulated by substrate stiffness in vitro while silencing blocked vascular development in vivo, and compromised barrier function in vitro. Together, SPTBN1 is identified as a new potential regulator of the leaky phenotype of atherosclerotic plaque microvessels.


Assuntos
Microvasos , Placa Aterosclerótica , Espectrina , Peixe-Zebra , Animais , Humanos , Permeabilidade Capilar , Células Endoteliais da Veia Umbilical Humana/metabolismo , Microvasos/patologia , Microvasos/metabolismo , Fenótipo , Placa Aterosclerótica/patologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Espectrina/genética , Espectrina/metabolismo , Transcriptoma , Peixe-Zebra/genética
10.
Angiogenesis ; 27(3): 561-582, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38775849

RESUMO

Coronary microvascular disease (CMD) and its progression towards major adverse coronary events pose a significant health challenge. Accurate in vitro investigation of CMD requires a robust cell model that faithfully represents the cells within the cardiac microvasculature. Human pluripotent stem cell-derived endothelial cells (hPSC-ECs) offer great potential; however, they are traditionally derived via differentiation protocols that are not readily scalable and are not specified towards the microvasculature. Here, we report the development and comprehensive characterisation of a scalable 3D protocol enabling the generation of phenotypically stable cardiac hPSC-microvascular-like ECs (hPSC-CMVECs) and cardiac pericyte-like cells. These were derived by growing vascular organoids within 3D stirred tank bioreactors and subjecting the emerging 3D hPSC-ECs to high-concentration VEGF-A treatment (3DV). Not only did this promote phenotypic stability of the 3DV hPSC-ECs; single cell-RNA sequencing (scRNA-seq) revealed the pronounced expression of cardiac endothelial- and microvascular-associated genes. Further, the generated mural cells attained from the vascular organoid exhibited markers characteristic of cardiac pericytes. Thus, we present a suitable cell model for investigating the cardiac microvasculature as well as the endothelial-dependent and -independent mechanisms of CMD. Moreover, owing to their phenotypic stability, cardiac specificity, and high angiogenic potential, the cells described within would also be well suited for cardiac tissue engineering applications.


Assuntos
Diferenciação Celular , Células Endoteliais , Microvasos , Células-Tronco Pluripotentes , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Microvasos/citologia , Microvasos/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Organoides/citologia , Organoides/irrigação sanguínea , Organoides/metabolismo
11.
Mol Med ; 30(1): 132, 2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39187765

RESUMO

Cell-based therapeutic strategies have been proposed as an alternative for brain and blood vessels repair after stroke, but their clinical application is hampered by potential adverse effects. We therefore tested the hypothesis that secretome of these cells might be used instead to still focus on cell-based therapeutic strategies. We therefore characterized the composition and the effect of the secretome of brain microvascular endothelial cells (BMECs) on primary in vitro human models of angiogenesis and vascular barrier. Two different secretome batches produced in high scale (scHSP) were analysed by mass spectrometry. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used as well as in vitro models of EC monolayer (CMECs) and blood-brain barrier (BBB). Cells were also exposed to oxygen-glucose deprivation (OGD) conditions and treated with scHSP during reoxygenation. Protein yield and composition of scHSP batches showed good reproducibility. scHSP increased CD34+-EC proliferation, tubulogenesis, and migration. Proteomic analysis of scHSP revealed the presence of growth factors and proteins modulating cell metabolism and inflammatory pathways. scHSP improved the integrity of CMECs, and upregulated the expression of junctional proteins. Such effects were mediated through the activation of the interferon pathway and downregulation of Wnt signalling. Furthermore, OGD altered the permeability of both CMECs and BBB, while scHSP prevented the OGD-induced vascular leakage in both models. These effects were mediated through upregulation of junctional proteins and regulation of MAPK/VEGFR2. Finally, our results highlight the possibility of using secretome from BMECs as a therapeutic alternative to promote brain angiogenesis and to protect from ischemia-induced vascular leakage.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Proteômica , Humanos , Células Endoteliais/metabolismo , Barreira Hematoencefálica/metabolismo , Proteômica/métodos , Secretoma/metabolismo , Permeabilidade Capilar , Encéfalo/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Hipóxia Celular , Proteoma/metabolismo , Células Cultivadas , Microvasos/metabolismo , Microvasos/citologia
12.
Am J Physiol Heart Circ Physiol ; 327(4): H1004-H1015, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39212765

RESUMO

Hypertension is associated with decreased endothelial function through reduced contributions of nitric oxide (NO). We previously discovered that flow-induced NO production in resistance arteries of mice and humans critically depends on endothelial inwardly rectifying K+ (Kir2.1) channels. The goal of this study was to establish whether these channels contribute to the impairment of endothelial function, measured by flow-induced vasodilation (FIV) in peripheral resistance arteries of humans with hypertension. We measured FIV in vessels isolated from subcutaneous fat biopsies from 32 subjects: normotensive [n = 19; 30.6 ± 9.8 yr old; systolic blood pressure (SBP): 115.2 ± 7 mmHg; diastolic blood pressure (DBP): 75.3 ± 5.7 mmHg] and hypertensive (n = 13; 45.3 ± 15.3 yr old; SBP: 146.1 ± 15.2 mmHg; DBP: 94.4 ± 6.9 mmHg). Consistent with previous studies, we find that FIV is impaired in hypertensive adults as demonstrated by a significant reduction in FIV when compared with the normotensive adults. Furthermore, our data suggest that the impairment of FIV in hypertensive adults is partially attributed to a reduction in Kir2.1-dependent vasodilation. Specifically, we show that blocking Kir2.1 with ML133 or functionally downregulating Kir2.1 with endothelial-specific adenoviral vector containing dominant-negative Kir2.1 (dnKir2.1) result in a significant reduction in FIV in normotensive subjects but with a smaller effect in hypertensive adults. The Kir2.1-dependent vasodilation was negatively correlated to both SBP and DBP, indicating that the Kir2.1 contribution to FIV decreases as blood pressure increases. In addition, we show that exposing vessels from normotensive adults to acute high-pressure results in loss of Kir2.1 contribution, as high pressure impairs vasodilation. No effect is seen when these vessels were incubated with dnKir2.1. Overexpressing wtKir2.1 in the endothelium resulted in some improvement in vasodilation in arteries from all participants, with a greater recovery in hypertensive adults. Our data suggest that hypertension-induced suppression of Kir2.1 is an important mechanism underlying endothelial dysfunction in hypertension.NEW & NOTEWORTHY Impairment of endothelial function under high blood pressure is linked to the loss of inwardly rectifying K+ (Kir2.1) channels activity in human resistance arteries, leading to a reduction in flow-induced vasodilation and possibly leading to a vicious cycle between elevation of blood pressure, and further impairment of Kir2.1 function and flow-induced vasodilation.


Assuntos
Endotélio Vascular , Hipertensão , Canais de Potássio Corretores do Fluxo de Internalização , Vasodilatação , Humanos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Hipertensão/fisiopatologia , Hipertensão/metabolismo , Hipertensão/genética , Pessoa de Meia-Idade , Masculino , Feminino , Adulto , Endotélio Vascular/fisiopatologia , Endotélio Vascular/metabolismo , Estudos de Casos e Controles , Pressão Sanguínea , Microvasos/fisiopatologia , Microvasos/metabolismo , Adulto Jovem , Células Endoteliais/metabolismo , Óxido Nítrico/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 327(1): H268-H274, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38787380

RESUMO

Brachial artery flow-mediated dilation (BAFMD) is induced by hyperemic wall shear rate (WSR) following forearm ischemia. In older adults, there appears to be a reduced brachial hyperemic WSR and altered stimulus-response relationship compared with young adults. However, it is unclear if an altered forearm microvascular response to ischemia influences brachial hyperemic WSR in older adults. We determined associations between brachial hyperemic WSR and forearm skeletal muscle oxygen saturation in young and older adults. Healthy young (n = 17, 29 ± 7 yr) and older (n = 32, 65 ± 4 yr) adults participated in the study. BAFMD by a multigate spectral Doppler system and forearm skeletal muscle oxygen saturation by near-infrared spectroscopy were concurrently measured. When compared with the young, older adults showed reduced oxygen extraction kinetics (OE, 0.15 [0.12-0.17] vs. 0.09 [0.05-0.12]%s-1) and magnitude (So2deficit, 3,810 ± 1,420 vs. 2,723 ± 1,240%s) during ischemia, as well as oxygen resaturation kinetics (So2slope, 2.5 ± 0.7 vs. 1.7 ± 0.7%s-1) upon reperfusion (all P < 0.05). When OE in the young and So2slope in older adults were stratified by their median values, young adults with OE above the median had greater hyperemic WSR parameters compared with those below the median (P < 0.05), but So2slope in older adults did not show clear differences in hyperemic WSR parameters between those above/below the median. This study demonstrates that, in addition to a reduced microvascular response to ischemia, there may be a dissociation between microvascular response to ischemia and brachial hyperemic WSR in older adults, which may result in a further impairment of BAFMD in this cohort.NEW & NOTEWORTHY Microvascular response to ischemia and subsequent reperfusion is diminished in older adults compared with the young. Furthermore, there appears to be a dissociation between the microvascular response to ischemia and brachial hyperemic WSR in older adults, which may further disturb the BAFMD process in this cohort. A reduced BAFMD in older adults may be a result of multiple alterations occurring both at macro- and microcirculation.


Assuntos
Artéria Braquial , Antebraço , Hiperemia , Microcirculação , Músculo Esquelético , Fluxo Sanguíneo Regional , Vasodilatação , Humanos , Artéria Braquial/fisiopatologia , Artéria Braquial/diagnóstico por imagem , Masculino , Feminino , Adulto , Idoso , Hiperemia/fisiopatologia , Hiperemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Pessoa de Meia-Idade , Antebraço/irrigação sanguínea , Adulto Jovem , Isquemia/fisiopatologia , Isquemia/metabolismo , Fatores Etários , Velocidade do Fluxo Sanguíneo , Espectroscopia de Luz Próxima ao Infravermelho , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Consumo de Oxigênio , Saturação de Oxigênio , Microvasos/fisiopatologia , Microvasos/metabolismo , Microvasos/diagnóstico por imagem
14.
Am J Physiol Heart Circ Physiol ; 327(4): H793-H803, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39058435

RESUMO

Women with a history of gestational diabetes mellitus (GDM) have a significantly greater lifetime risk of developing cardiovascular disease and type 2 diabetes compared with women who had an uncomplicated pregnancy (HC). Microvascular endothelial dysfunction, mediated via reduced nitric oxide (NO)-dependent dilation secondary to increases in oxidative stress, persists after pregnancy complicated by GDM. We examined whether this microvascular dysfunction reduces insulin-mediated vascular responses in women with a history of GDM. We assessed in vivo microvascular endothelium-dependent vasodilator function by measuring cutaneous vascular conductance responses to graded infusions of acetylcholine (10-10-10-1 M) and insulin (10-8-10-4 M) in control sites and sites treated with 15 mM l-NAME [NG-nitro-l-arginine methyl ester; NO-synthase (NOS) inhibitor] or 5 mM l-ascorbate. We also measured protein expression of total endothelial NOS (eNOS), insulin-mediated eNOS phosphorylation, and endothelial nitrotyrosine in isolated endothelial cells from GDM and HC. Women with a history of GDM had reduced acetylcholine (P < 0.001)- and insulin (P < 0.001)-mediated dilation, and the NO-dependent responses to both acetylcholine (P = 0.006) and insulin (P = 0.006) were reduced in GDM compared with HC. Insulin stimulation increased phosphorylated eNOS content in HC (P = 0.009) but had no effect in GDM (P = 0.306). Ascorbate treatment increased acetylcholine (P < 0.001)- and insulin (P < 0.001)-mediated dilation in GDM, and endothelial cell nitrotyrosine expression was higher in GDM compared with HC (P = 0.014). Women with a history of GDM have attenuated microvascular vasodilation responses to insulin, and this attenuation is mediated, in part, by reduced NO-dependent mechanisms. Our findings further implicate increased endothelial oxidative stress in this microvascular insulin resistance.NEW & NOTEWORTHY Women who have gestational diabetes during pregnancy are at a greater risk for cardiovascular disease and type 2 diabetes in the decade following pregnancy. The mechanisms mediating this increased risk are unclear. Herein, we demonstrate that insulin-dependent microvascular responses are reduced in women who had gestational diabetes, despite the remission of glucose intolerance. This reduced microvascular sensitivity to insulin may contribute to increased cardiovascular disease and type 2 diabetes risk in these women.


Assuntos
Diabetes Gestacional , Insulina , Microvasos , Óxido Nítrico Sintase Tipo III , Óxido Nítrico , Vasodilatação , Feminino , Diabetes Gestacional/fisiopatologia , Diabetes Gestacional/metabolismo , Humanos , Gravidez , Vasodilatação/efeitos dos fármacos , Insulina/farmacologia , Adulto , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Microvasos/metabolismo , Microvasos/efeitos dos fármacos , Microvasos/fisiopatologia , Acetilcolina/farmacologia , Vasodilatadores/farmacologia , Fosforilação , Estresse Oxidativo/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Endotélio Vascular/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/metabolismo , Estudos de Casos e Controles , NG-Nitroarginina Metil Éster/farmacologia , Pele/irrigação sanguínea
15.
Am J Physiol Heart Circ Physiol ; 327(2): H364-H369, 2024 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-38847757

RESUMO

The transcriptional regulator nuclear factor-κB (NF-κB) is a mediator of endothelial dysfunction. Inhibiting NF-κB with salsalate is used to investigate inflammatory mechanisms contributing to accelerated cardiovascular disease risk. However, in the absence of disease, inhibition of NF-κB can impact redox mechanisms, resulting in paradoxically decreased endothelial function. This study aimed to measure microvascular endothelial function during inhibition of the transcriptional regulator NF-κB in reproductive-aged healthy women. In a randomized, single-blind, crossover, placebo-controlled design, nine healthy women were randomly assigned oral salsalate (1,500 mg, twice daily) or placebo treatments for 5 days. Subjects underwent graded perfusion with the endothelium-dependent agonist acetylcholine (ACh, 10-10 to 10-1 M, 33°C) alone and in combination with 15 mM NG-nitro-l-arginine methyl ester [l-NAME; nonselective nitric oxide (NO) synthase inhibitor] through intradermal microdialysis. Laser-Doppler flux was measured over each microdialysis site, and cutaneous vascular conductance (CVC) was calculated as flux divided by mean arterial pressure and normalized to site-specific maximum (CVC%max; 28 mM sodium nitroprusside + 43°C). The l-NAME sensitive component was calculated as the difference between the areas under the dose-response curves. During the placebo and salsalate treatments, the l-NAME sites were reduced compared with the control sites (both P < 0.0001). Across treatments, there was a significant difference between the control and l-NAME sites, where both sites shifted upward following salsalate treatment (both P < 0.0001), whereas the l-NAME-sensitive component was not different (P = 0.94). These data demonstrate that inhibition of the transcriptional regulator NF-κB improves cutaneous microvascular function in reproductive-aged healthy women through non-NO-dependent mechanisms.NEW & NOTEWORTHY The transcription factor nuclear factor-κB (NF-κB) regulates multiple aspects of innate and adaptive immunity by encoding for genes that participate in inflammation and impact endothelial function following NF-κB inhibition with salsalate treatment. Our results show that cutaneous microvascular function is increased through non-nitric oxide (NO)-dependent mechanisms following salsalate treatment in reproductive-aged healthy women.


Assuntos
Estudos Cross-Over , Microcirculação , NF-kappa B , Óxido Nítrico , Pele , Humanos , Feminino , Adulto , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Pele/metabolismo , NF-kappa B/metabolismo , Método Simples-Cego , Microcirculação/efeitos dos fármacos , Óxido Nítrico/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Vasodilatação/efeitos dos fármacos , Adulto Jovem , Acetilcolina/farmacologia , Voluntários Saudáveis , Vasodilatadores/farmacologia , Inibidores Enzimáticos/farmacologia , Salicilatos/farmacologia , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos
16.
Biochem Biophys Res Commun ; 724: 150234, 2024 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-38865812

RESUMO

Vasculature-on-chip (VoC) models have become a prominent tool in the study of microvasculature functions because of their cost-effective and ethical production process. These models typically use a hydrogel in which the three-dimensional (3D) microvascular structure is embedded. Thus, VoCs are directly impacted by the physical and chemical cues of the supporting hydrogel. Endothelial cell (EC) response in VoCs is critical, especially in organ-specific vasculature models, in which ECs exhibit specific traits and behaviors that vary between organs. Many studies customize the stimuli ECs perceive in different ways; however, customizing the hydrogel composition accordingly to the target organ's extracellular matrix (ECM), which we believe has great potential, has been rarely investigated. We explored this approach to organ-specific VoCs by fabricating microvessels (MVs) with either human umbilical vein ECs or human brain microvascular ECs in a 3D cylindrical VoC using a collagen hydrogel alone or one supplemented with laminin and hyaluronan, components found in the brain ECM. We characterized the physical properties of these hydrogels and analyzed the barrier properties of the MVs. Barrier function and tight junction (ZO-1) expression improved with the addition of laminin and hyaluronan in the composite hydrogel.


Assuntos
Colágeno , Células Endoteliais da Veia Umbilical Humana , Ácido Hialurônico , Hidrogéis , Laminina , Microvasos , Junções Íntimas , Humanos , Hidrogéis/química , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Laminina/química , Laminina/metabolismo , Colágeno/química , Colágeno/metabolismo , Microvasos/metabolismo , Microvasos/efeitos dos fármacos , Junções Íntimas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Dispositivos Lab-On-A-Chip , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Cultivadas
17.
Microcirculation ; 31(7): e12877, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39222475

RESUMO

The brain microvasculature, which delivers oxygen and nutrients and forms a critical barrier protecting the central nervous system via capillaries, is deleteriously affected by both Alzheimer's disease (AD) and type 2 diabetes (T2D). T2D patients have an increased risk of developing AD, suggesting potentially related microvascular pathological mechanisms. Pericytes are an ideal cell type to study for functional links between AD and T2D. These specialized capillary-enwrapping cells regulate capillary density, lumen diameter, and blood flow. Pericytes also maintain endothelial tight junctions to ensure blood-brain barrier integrity, modulation of immune cell extravasation, and clearance of toxins. Changes in these phenomena have been observed in both AD and T2D, implicating "pericyte pathology" as a common feature of AD and T2D. This review examines the mechanisms of AD and T2D from the perspective of the brain microvasculature, highlighting how pericyte pathology contributes to both diseases. Our review identifies voids in understanding how AD and T2D negatively impact the brain microvasculature and suggests future studies to examine the intersections of these diseases.


Assuntos
Doença de Alzheimer , Barreira Hematoencefálica , Encéfalo , Diabetes Mellitus Tipo 2 , Microvasos , Pericitos , Pericitos/patologia , Pericitos/metabolismo , Humanos , Doença de Alzheimer/patologia , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Encéfalo/patologia , Encéfalo/irrigação sanguínea , Microvasos/patologia , Microvasos/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/metabolismo , Animais
18.
Microcirculation ; 31(5): e12859, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38818977

RESUMO

OBJECTIVE: The endothelium regulates crucial aspects of vascular function, including hemostasis, vasomotor tone, proliferation, immune cell adhesion, and microvascular permeability. Endothelial cells (ECs), especially in arterioles, are pivotal for flow distribution and peripheral resistance regulation. Investigating vascular endothelium physiology, particularly in microvascular ECs, demands precise isolation and culturing techniques. METHODS: Freshly isolated ECs are vital for examining protein expression, ion channel behavior, and calcium dynamics. Establishing primary endothelial cell cultures is crucial for unraveling vascular functions and understanding intact microvessel endothelium roles. Despite the significance, detailed protocols and comparisons with intact vessels are scarce in microvascular research. We developed a reproducible method to isolate microvascular ECs, assessing substrate influence by cultivating cells on fibronectin and gelatin matrix gels. This comparative approach enhances our understanding of microvascular endothelial cell biology. RESULTS: Microvascular mesenteric ECs expressed key markers (VE-cadherin and eNOS) in both matrix gels, confirming cell culture purity. Under uncoated conditions, ECs were undetected, whereas proteins linked to smooth muscle cells and fibroblasts were evident. Examining endothelial cell (EC) physiological dynamics on distinct matrix substrates revealed comparable cell length, shape, and Ca2+ elevations in both male and female ECs on gelatin and fibronectin matrix gels. Gelatin-cultured ECs exhibited analogous membrane potential responses to acetylcholine (ACh) or adenosine triphosphate (ATP), contrasting with their fibronectin-cultured counterparts. In the absence of stimulation, fibronectin-cultured ECs displayed a more depolarized resting membrane potential than gelatin-cultured ECs. CONCLUSIONS: Gelatin-cultured ECs demonstrated electrical behaviors akin to intact endothelium from mouse mesenteric arteries, thus advancing our understanding of endothelial cell behavior within diverse microenvironments.


Assuntos
Células Endoteliais , Gelatina , Microvasos , Óxido Nítrico Sintase Tipo III , Animais , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Camundongos , Feminino , Masculino , Microvasos/citologia , Microvasos/metabolismo , Microvasos/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Células Cultivadas , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Géis , Antígenos CD/metabolismo , Caderinas/metabolismo , Cultura Primária de Células , Endotélio Vascular/metabolismo , Endotélio Vascular/citologia
19.
Microcirculation ; 31(7): e12876, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39005221

RESUMO

OBJECTIVE: Abnormal tumor vascular network contributes to aberrant blood perfusion and reduced oxygenation in tumors, which lead to poor efficacy of chemotherapy and radiotherapy. We aimed to explore the effects of the tumor-derived exosomes (TDEs) and C188-9 (a small molecule inhibitor of signal transducer and activator of transcription 3, STAT3) on tumor microvascular hemodynamics and determine which blood flow oscillations for various frequency intervals are responsible for these changes. METHODS: Microvascular hemodynamics parameters were recorded using a PeriFlux 6000 EPOS system in tumor surface in a nude mouse subcutaneous xenograft model. Oscillations of laser Doppler flowmetry (LDF) signal were investigated by wavelet transform analysis. RESULTS: TDEs facilitated tumor growth at least partially was associated with increasing blood flow in smaller vessels with lower speed and decreasing the blood flow at larger vessels with higher speed. Lower oxyhemoglobin saturation (SO2) on tumor surface was aggravated by TDEs, and C188-9 treatment significantly alleviated this decrease. Wavelet transform spectral analysis revealed that TDEs increased the amplitude of oscillations in four frequency intervals related to endothelial (NO-dependent and -independent), myogenic and neurogenic activities, and C188-9 had no effect on this increase. CONCLUSIONS: TDEs facilitated tumor growth partially was associated with increasing blood flow in distributing vessels, reducing blood perfusion in larger vessels, and lowering SO2 on tumor surface. Enhanced vascular smooth muscle, endothelial and neurogenic activities occurred in tumor superficial zone.


Assuntos
Exossomos , Neoplasias Ovarianas , Animais , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/fisiopatologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/irrigação sanguínea , Humanos , Camundongos , Exossomos/metabolismo , Feminino , Camundongos Nus , Hemodinâmica , Linhagem Celular Tumoral , Microvasos/fisiopatologia , Microvasos/metabolismo , Microcirculação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Xenoenxertos , Neovascularização Patológica/metabolismo , Fator de Transcrição STAT3/metabolismo
20.
J Bioenerg Biomembr ; 56(3): 193-204, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38446318

RESUMO

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.


Assuntos
Monoterpenos Acíclicos , Apoptose , Células Endoteliais , Glucose , Heme Oxigenase-1 , Fator 2 Relacionado a NF-E2 , Espécies Reativas de Oxigênio , Humanos , Apoptose/efeitos dos fármacos , Monoterpenos Acíclicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Glucose/metabolismo , Heme Oxigenase-1/metabolismo , Oxigênio/metabolismo , Encéfalo/metabolismo , Encéfalo/irrigação sanguínea , Microvasos/metabolismo , Microvasos/patologia , Microvasos/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA