Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.545
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 325(1): H172-H186, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37294893

RESUMO

The adipokine chemerin may support blood pressure, evidenced by a fall in mean arterial pressure after whole body antisense oligonucleotide (ASO)-mediated knockdown of chemerin protein in rat models of normal and elevated blood pressure. Although the liver is the greatest contributor of circulating chemerin, liver-specific ASOs that abolished hepatic-derived chemerin did not change blood pressure. Thus, other sites must produce the chemerin that supports blood pressure. We hypothesize that the vasculature is a source of chemerin independent of the liver that supports arterial tone. RNAScope, PCR, Western blot analyses, ASOs, isometric contractility, and radiotelemetry were used in the Dahl salt-sensitive (SS) rat (male and female) on a normal diet. Retinoic acid receptor responder 2 (Rarres2) mRNA was detected in the smooth muscle, adventitia, and perivascular adipose tissue of the thoracic aorta. Chemerin protein was detected immunohistochemically in the endothelium, smooth muscle cells, adventitia, and perivascular adipose tissue. Chemerin colocalized with the vascular smooth muscle marker α-actin and the adipocyte marker perilipin. Importantly, chemerin protein in the thoracic aorta was not reduced when liver-derived chemerin was abolished by a liver-specific ASO against chemerin. Chemerin protein was similarly absent in arteries from a newly created global chemerin knockout in Dahl SS rats. Inhibition of the receptor Chemerin1 by the receptor antagonist CCX832 resulted in the loss of vascular tone that supports potential contributions of chemerin by both perivascular adipose tissue and the media. These data suggest that vessel-derived chemerin may support vascular tone locally through constitutive activation of Chemerin1. This posits chemerin as a potential therapeutic target in blood pressure regulation.NEW & NOTEWORTHY Vascular tunicas synthesizing chemerin is a new finding. Vascular chemerin is independent of hepatic-derived chemerin. Vasculature from both males and females have resident chemerin. Chemerin1 receptor activity supports vascular tone.


Assuntos
Vasos Sanguíneos , Quimiocinas , Animais , Ratos , Técnicas de Silenciamento de Genes , Fígado/metabolismo , Aorta/metabolismo , Quimiocinas/análise , Quimiocinas/metabolismo , Músculo Liso Vascular/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia
2.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-36675082

RESUMO

A number of stressors and inflammatory mediators (cytokines, proteases, oxidative stress mediators) released during inflammation or ischemia stimulate and activate cells in blood, the vessel wall or tissues. The most well-known functional and phenotypic responses of activated cells are (1) the immediate expression and/or release of stored or newly synthesized bioactive molecules, and (2) membrane blebbing followed by release of microvesicles. An ultimate response, namely the formation of extracellular traps by neutrophils (NETs), is outside the scope of this work. The main objective of this article is to provide an overview on the mechanism of plasminogen reception and activation at the surface of cell-derived microvesicles, new actors in fibrinolysis and proteolysis. The role of microvesicle-bound plasmin in pathological settings involving inflammation, atherosclerosis, angiogenesis, and tumour growth, remains to be investigated. Further studies are necessary to determine if profibrinolytic microvesicles are involved in a finely regulated equilibrium with pro-coagulant microvesicles, which ensures a balanced haemostasis, leading to the maintenance of vascular patency.


Assuntos
Micropartículas Derivadas de Células , Fibrinólise , Plasminogênio , Proteólise , Humanos , Vasos Sanguíneos/metabolismo , Fibrinolisina/metabolismo , Fibrinólise/fisiologia , Inflamação/metabolismo , Plasminogênio/metabolismo , Micropartículas Derivadas de Células/metabolismo
3.
Methods Mol Biol ; 2572: 55-66, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36161407

RESUMO

Tumor vasculature is the major extrinsic factor that shapes Intra-tumoral heterogeneity (ITH). Non-uniform exposure of microenvironmental cues greatly impacts cancer cell phenotypes leading to ITH, which exacerbates therapy resistance. This raises a need to study the influence of non-uniform perfusion patterns and the resulting heterogeneity that persists within the tumor microenvironment (TME). A method was developed to identify cancer cells based on their proximity to functional blood vessels (BVs) called perfusion-based fluorescent dye labeling of cells (PFDLC). PFDLC works on the principle of perfusion, where a freely diffusible nuclear binding fluorescent dye (Hoechst 33342) is injected intravenously (i.v.) through a tail vein into atumor-bearing mice. The tumors are retrieved post dye perfusion, dissociated into single cells, and sorted based on their dye uptake proportional to their distance from the nearest blood capillary. This method is amenable to multi-omics as well as functional assays.


Assuntos
Corantes Fluorescentes , Neoplasias , Animais , Vasos Sanguíneos/metabolismo , Movimento Celular , Corantes Fluorescentes/metabolismo , Camundongos , Neoplasias/metabolismo , Perfusão , Microambiente Tumoral
4.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36232721

RESUMO

During vascular development, endothelial cAMP-dependent protein kinase A (PKA) regulates angiogenesis by controlling the number of tip cells, and PKA inhibition leads to excessive angiogenesis. Whether this role of endothelial PKA is restricted to embryonic and neonatal development or is also required for vascular homeostasis later on is unknown. Here, we show that perinatal (postnatal days P1-P3) of later (P28-P32) inhibition of endothelial PKA using dominant-negative PKA expressed under the control of endothelial-specific Cdh5-CreERT2 recombinase (dnPKAiEC mice) leads to severe subcutaneous edema, hypoalbuminemia, hypoglycemia and premature death. These changes were accompanied by the local hypersprouting of blood vessels in fat pads and the secondary enlargement of subcutaneous lymphatic vessels. Most noticeably, endothelial PKA inhibition caused a dramatic disorganization of the liver vasculature. Hepatic changes correlated with decreased gluconeogenesis, while liver albumin production seems to be unaffected and hypoalbuminemia is rather a result of increased leakage into the interstitium. Interestingly, the expression of dnPKA only in lymphatics using Prox1-CreERT2 produced no phenotype. Likewise, the mosaic expression in only endothelial subpopulations using Vegfr3-CreERT2 was insufficient to induce edema or hypoglycemia. Increased expression of the tip cell marker ESM1 indicated that the inhibition of PKA induced an angiogenic response in the liver, although tissue derived pro- and anti-angiogenic factors were unchanged. These data indicate that endothelial PKA is a gatekeeper of endothelial cell activation not only in development but also in adult homeostasis, preventing the aberrant reactivation of the angiogenic program.


Assuntos
Vasos Sanguíneos , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico , Células Endoteliais , Fígado , Albuminas , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , AMP Cíclico , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Homeostase , Hipoalbuminemia , Hipoglicemia , Fígado/metabolismo , Fígado/fisiologia , Camundongos , Recombinases
5.
Dev Cell ; 57(6): 767-782.e6, 2022 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-35276066

RESUMO

During embryogenesis, the initial vascular network forms by the process of vasculogenesis, or the specification of vascular progenitors de novo. In contrast, the majority of later-forming vessels arise by angiogenesis from the already established vasculature. Here, we show that new vascular progenitors in zebrafish embryos emerge from a distinct site along the yolk extension, or secondary vascular field (SVF), incorporate into the posterior cardinal vein, and contribute to subintestinal vasculature even after blood circulation has been initiated. We further demonstrate that SVF cells participate in vascular recovery after chemical ablation of vascular endothelial cells. Inducible inhibition of the function of vascular progenitor marker etv2/etsrp prevented SVF cell differentiation and resulted in the defective formation of subintestinal vasculature. Similar late-forming etv2+ progenitors were also observed in mouse embryos, suggesting that SVF cells are evolutionarily conserved. Our results characterize a distinct mechanism by which new vascular progenitors incorporate into established vasculature.


Assuntos
Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Vasos Sanguíneos/metabolismo , Embrião não Mamífero/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Neovascularização Fisiológica , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
6.
Nat Metab ; 4(1): 123-140, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35102339

RESUMO

Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Matriz Extracelular/metabolismo , Fosforilação Oxidativa , Via de Pentose Fosfato , Animais , Biomarcadores , Elastina/biossíntese , Elastina/genética , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Expressão Gênica , Genes Reporter , Glucose/metabolismo , Hemodinâmica , Camundongos , Camundongos Knockout , Modelos Biológicos , Estresse Oxidativo , Pentosefosfatos/metabolismo , Peixe-Zebra
7.
Molecules ; 27(4)2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35209016

RESUMO

Hot flashes are considered the most bothersome complaint during menopause. Although hormone therapy is an effective option to relieve hot flashes, it has been associated with significant side effects. The aim of our study is to suggest a novel combination of different plant extracts with distinct mechanisms of action against hot flashes. We selected the rhizome of Glycyrrhiza glabra L. (Fabaceae), the rhizome of Actaea racemosa L. (Ranunculaceae), the aerial parts of Hypericum perforatum L. (Hypericaceae) to produce extracts rich in bioactive phytochemicals and the seed oil of Oenothera biennis L. (Onagraceae). We investigated their estrogenic and antioxidant potential and their inhibitory effect against prostaglandin D2 receptor 1 (DP1) as a novel mechanistic pathway for vasodilation in hot flashes, alone or in combination. The phytochemical footprint of the extracts was analyzed using HPLC-PDA and UPLC-HRMS. We observed that the tested extracts possess different mechanisms of action. A. racemosa exerts a beneficial activation of the estrogen receptor, H. perforatum possesses the highest antioxidant capacity and the seed oil of O. biennis inhibits the DP1 receptor. The triple combination in the optimal doses pertains to efficacy against all three mechanisms of action, serves as a multitarget plant-based therapy and could serve as a novel strategy for the alleviation of hot flashes in postmenopausal women.


Assuntos
Fogachos/tratamento farmacológico , Menopausa , Extratos Vegetais/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Linhagem Celular Tumoral , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Estrogênios/química , Estrogênios/farmacologia , Humanos , Menopausa/efeitos dos fármacos , Pessoa de Meia-Idade , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacologia , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Prostaglandinas/metabolismo
8.
Int J Mol Sci ; 23(4)2022 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-35216218

RESUMO

Many extensible tissues such as skin, lungs, and blood vessels require elasticity to function properly. The recoil of elastic energy stored during a stretching phase is provided by elastic fibers, which are mostly composed of elastin and fibrillin-rich microfibrils. In arteries, the lack of elastic fibers leads to a weakening of the vessel wall with an increased risk to develop cardiovascular defects such as stenosis, aneurysms, and dissections. The development of new therapeutic molecules involves preliminary tests in animal models that recapitulate the disease and whose response to drugs should be as close as possible to that of humans. Due to its superior in vivo imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study human pathologies. Moreover, it is particularly adapted to large scale studies, making it an attractive model in particular for the first steps of investigations. In this review, we discuss the relevance of the zebrafish model for the study of elastic fiber-related vascular pathologies. We evidence zebrafish as a compelling alternative to conventional mouse models.


Assuntos
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Tecido Elástico/metabolismo , Tecido Elástico/patologia , Fibrilinas/metabolismo , Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Elasticidade/fisiologia , Humanos , Proteínas dos Microfilamentos/metabolismo
9.
Nat Commun ; 13(1): 215, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017526

RESUMO

Macrophages are integral to the pathogenesis of atherosclerosis, but the contribution of distinct macrophage subsets to disease remains poorly defined. Using single cell technologies and conditional ablation via a LysMCre+ Clec4a2flox/DTR mouse strain, we demonstrate that the expression of the C-type lectin receptor CLEC4A2 is a distinguishing feature of vascular resident macrophages endowed with athero-protective properties. Through genetic deletion and competitive bone marrow chimera experiments, we identify CLEC4A2 as an intrinsic regulator of macrophage tissue adaptation by promoting a bias in monocyte-to-macrophage in situ differentiation towards colony stimulating factor 1 (CSF1) in vascular health and disease. During atherogenesis, CLEC4A2 deficiency results in loss of resident vascular macrophages and their homeostatic properties causing dysfunctional cholesterol metabolism and enhanced toll-like receptor triggering, exacerbating disease. Our study demonstrates that CLEC4A2 licenses monocytes to join the vascular resident macrophage pool, and that CLEC4A2-mediated macrophage homeostasis is critical to combat cardiovascular disease.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Vasos Sanguíneos/metabolismo , Lectinas Tipo C/genética , Macrófagos/metabolismo , Animais , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Vasos Sanguíneos/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Morte Celular/genética , Diferenciação Celular , Linhagem da Célula/genética , Colesterol/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Homeostase/genética , Humanos , Lectinas Tipo C/deficiência , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Transdução de Sinais , Análise de Célula Única
11.
Circ Res ; 130(3): 366-383, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986653

RESUMO

BACKGROUND: The chromatin-remodeling enzyme BRG1 (brahma-related gene 1) regulates gene expression in a variety of rapidly differentiating cells during embryonic development. However, the critical genes that BRG1 regulates during lymphatic vascular development are unknown. METHODS: We used genetic and imaging techniques to define the role of BRG1 in murine embryonic lymphatic development, although this approach inadvertently expanded our study to multiple interacting cell types. RESULTS: We found that omental macrophages fine-tune an unexpected developmental process by which erythrocytes escaping from naturally discontinuous omental blood vessels are collected by nearby lymphatic vessels. Our data indicate that circulating fibrin(ogen) leaking from gaps in omental blood vessels can trigger inflammasome-mediated IL-1ß (interleukin-1ß) production and secretion from nearby macrophages. IL-1ß destabilizes adherens junctions in omental blood and lymphatic vessels, contributing to both extravasation of erythrocytes and their uptake by lymphatics. BRG1 regulates IL-1ß production in omental macrophages by transcriptionally suppressing the inflammasome trigger RIPK3 (receptor interacting protein kinase 3). CONCLUSIONS: Genetic deletion of Brg1 in embryonic macrophages leads to excessive IL-1ß production, erythrocyte leakage from blood vessels, and blood-filled lymphatics in the developing omentum. Altogether, these results highlight a novel context for epigenetically regulated crosstalk between macrophages, blood vessels, and lymphatics.


Assuntos
Vasos Sanguíneos/metabolismo , DNA Helicases/metabolismo , Interleucina-1beta/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Nucleares/metabolismo , Omento/metabolismo , Fatores de Transcrição/metabolismo , Junções Aderentes/metabolismo , Animais , Vasos Sanguíneos/embriologia , DNA Helicases/genética , Eritrócitos/metabolismo , Inflamassomos/metabolismo , Vasos Linfáticos/embriologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Omento/irrigação sanguínea , Omento/embriologia , Fatores de Transcrição/genética
12.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055054

RESUMO

Endothelial progenitor cells (EPCs) are involved in vascular repair and modulate properties of smooth muscle cells (SMCs) relevant for their contribution to neointima formation following injury. Considering the relevant role of the CXCL12-CXCR4 axis in vascular homeostasis and the potential of EPCs and SMCs to release CXCL12 and express CXCR4, we analyzed the engagement of the CXCL12-CXCR4 axis in various modes of EPC-SMC interaction relevant for injury- and lipid-induced atherosclerosis. We now demonstrate that the expression and release of CXCL12 is synergistically increased in a CXCR4-dependent mechanism following EPC-SMC interaction during co-cultivation or in response to recombinant CXCL12, thus establishing an amplifying feedback loop Additionally, mechanical injury of SMCs induces increased release of CXCL12, resulting in enhanced CXCR4-dependent recruitment of EPCs to SMCs. The CXCL12-CXCR4 axis is crucially engaged in the EPC-triggered augmentation of SMC migration and the attenuation of SMC apoptosis but not in the EPC-mediated increase in SMC proliferation. Compared to EPCs alone, the alliance of EPC-SMC is superior in promoting the CXCR4-dependent proliferation and migration of endothelial cells. When direct cell-cell contact is established, EPCs protect the contractile phenotype of SMCs via CXCL12-CXCR4 and reverse cholesterol-induced transdifferentiation toward a synthetic, macrophage-like phenotype. In conclusion we show that the interaction of EPCs and SMCs unleashes a CXCL12-CXCR4-based autoregulatory feedback loop promoting regenerative processes and mediating SMC phenotype control to potentially guard vascular homeostasis.


Assuntos
Vasos Sanguíneos/metabolismo , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliais/metabolismo , Homeostase , Miócitos de Músculo Liso/metabolismo , Receptores CXCR4/metabolismo , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Biomarcadores , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/genética , Expressão Gênica , Humanos , Neointima/genética , Neointima/metabolismo , Fenótipo , Ligação Proteica , Receptores CXCR4/genética , Transdução de Sinais
13.
Int J Mol Sci ; 23(2)2022 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-35055149

RESUMO

Vascular disease was for a long time considered a disease of the old age, but it is becoming increasingly clear that a cumulus of factors can cause early vascular aging (EVA). Inflammation plays a key role in vascular stiffening and also in other pathologies that induce vascular damage. There is a known and confirmed connection between inflammation and atherosclerosis. However, it has taken a long time to prove the beneficial effects of anti-inflammatory drugs on cardiovascular events. Diabetes can be both a product of inflammation and a cofactor implicated in the progression of vascular disease. When diabetes and inflammation are accompanied by obesity, this ominous trifecta leads to an increased incidence of atherothrombotic events. Research into earlier stages of vascular disease, and documentation of vulnerability to premature vascular disease, might be the key to success in preventing clinical events. Modulation of inflammation, combined with strict control of classical cardiovascular risk factors, seems to be the winning recipe. Identification of population subsets with a successful vascular aging (supernormal vascular aging-SUPERNOVA) pattern could also bring forth novel therapeutic interventions.


Assuntos
Envelhecimento/metabolismo , Aterosclerose/metabolismo , Vasos Sanguíneos/metabolismo , Diabetes Mellitus/metabolismo , Inflamação/metabolismo , Trombose/metabolismo , Animais , Aterosclerose/etiologia , Vasos Sanguíneos/efeitos dos fármacos , Diabetes Mellitus/etiologia , Humanos , Inflamação/complicações , Inflamação/tratamento farmacológico , Obesidade/etiologia , Obesidade/metabolismo , Trombose/etiologia
14.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166339, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35017029

RESUMO

The pancreatic islet vasculature is of fundamental importance to the ß-cell response to obesity-associated insulin resistance. To explore islet vascular alterations in the pathogenesis of type 2 diabetes, we evaluated two insulin resistance models: ob/ob mice, which sustain large ß-cell mass and hyperinsulinemia, and db/db mice, which progress to diabetes due to secondary ß-cell compensation failure for insulin secretion. Time-dependent changes in islet vasculature and blood flow were investigated using tomato lectin staining and in vivo live imaging. Marked islet capillary dilation was observed in ob/ob mice, but this adaptive change was blunted in db/db mice. Islet blood flow volume was augmented in ob/ob mice, whereas it was reduced in db/db mice. The protein concentrations of total and phosphorylated endothelial nitric oxide synthase (eNOS) at Ser1177 were increased in ob/ob islets, while they were diminished in db/db mice, indicating decreased eNOS activity. This was accompanied by an increased retention of advanced glycation end-products in db/db blood vessels. Amelioration of diabetes by Elovl6 deficiency involved a restoration of capillary dilation, blood flow, and eNOS phosphorylation in db/db islets. Our findings suggest that the disability of islet capillary dilation due to endothelial dysfunction impairs local islet blood flow, which may play a role in the loss of ß-cell function and further exacerbate type 2 diabetes.


Assuntos
Vasos Sanguíneos/metabolismo , Ilhotas Pancreáticas/fisiologia , Animais , Velocidade do Fluxo Sanguíneo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Elongases de Ácidos Graxos/deficiência , Elongases de Ácidos Graxos/genética , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Insulina/metabolismo , Resistência à Insulina , Ilhotas Pancreáticas/anatomia & histologia , Ilhotas Pancreáticas/irrigação sanguínea , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação
15.
Biochem Biophys Res Commun ; 592: 60-66, 2022 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-35033869

RESUMO

BACKGROUND AND AIMS: Atherosclerosis is driven by an inflammatory process of the vascular wall. The novel orphan G-protein coupled receptor 5B of family C (GPRC5B) is involved in drosophila sugar and lipid metabolism as well as mice adipose tissue inflammation. Here, we investigated the role of GPRC5B in the pro-atherogenic mechanisms of hyperglycemia and vascular inflammation. METHODS: Immortalized and primary endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) were used for stimulation with high glucose or different cytokines. Adenoviral- or plasmid-driven GPRC5B overexpression and siRNA-mediated knockdown were performed in these cells to analyze functional and mechanistic pathways of GPRC5B. RESULTS: In ECs and VSMCs, stimulation with high glucose, TNFα or LPS induced a significant upregulation of endogenous GPRC5B mRNA and protein levels. GPRC5B overexpression and knockdown increased and attenuated, respectively, the expression of the pro-inflammatory cytokines TNFα, IL-1ß, IL-6 as well as the pro-atherogenic vascular adhesion molecules ICAM-1 and VCAM-1. Furthermore, the expression and activity of the metalloproteinase MMP-9, a component of atherosclerotic plaque stabilization, were significantly enhanced by GPRC5B overexpression. Mechanistically, GPRC5B increased the phosphorylation of ERK1/2 and activated NFκB through a direct interaction with the tyrosine kinase Fyn. CONCLUSIONS: Our findings demonstrate that GPRC5B is upregulated in response to high glucose and pro-inflammatory signaling. GPRC5B functionally modulates the inflammatory activity in cells of the vascular wall, suggesting a pro-atherogenic GPRC5B-dependent positive feedback loop via Fyn and NFκB. Thus, GPRC5B warrants further attention as a novel pharmacological target for the treatment of vascular inflammation and possibly atherogenesis.


Assuntos
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Inflamação/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Vasos Sanguíneos/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Citocinas/efeitos adversos , Ativação Enzimática/efeitos dos fármacos , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hiperglicemia/patologia , Inflamação/patologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos
16.
Am J Physiol Heart Circ Physiol ; 322(1): H25-H35, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34738833

RESUMO

Central adiposity is associated with greater sympathetic support of blood pressure. ß-adrenergic receptors (ß-AR) buffer sympathetically mediated vasoconstriction and ß-AR-mediated vasodilation is attenuated in preclinical models of obesity. With this information, we hypothesized ß-AR vasodilation would be lower in obese compared with normal weight adults. Because ß-AR vasodilation in normal weight adults is limited by cyclooxygenase (COX) restraint of nitric oxide synthase (NOS), we further explored the contributions of COX and NOS to ß-AR vasodilation in this cohort. Forearm blood flow (FBF, Doppler ultrasound) and mean arterial blood pressure (MAP, brachial arterial catheter) were measured and forearm vascular conductance (FVC) was calculated (FVC = FBF/MAP). The rise in FVC from baseline (ΔFVC) was quantified during graded brachial artery infusion of isoproterenol (Iso, 1-12 ng/100 g/min) in normal weight (n = 36) and adults with obesity (n = 22) (18-40 yr old). In a subset of participants, Iso-mediated vasodilation was examined before and during inhibition of NOS [NG-monomethyl-l-arginine (l-NMMA)], COX (ketorolac), and NOS + COX (l-NMMA + ketorolac). Iso-mediated increases in FVC did not differ between groups (P = 0.57). l-NMMA attenuated Iso-mediated ΔFVC in normal weight (P = 0.03) but not adults with obesity (P = 0.27). In normal weight adults, ketorolac increased Iso-mediated ΔFVC (P < 0.01) and this response was lost with concurrent l-NMMA (P = 0.67). In contrast, neither ketorolac (P = 0.81) nor ketorolac + l-NMMA (P = 0.40) altered Iso-mediated ΔFVC in adults with obesity. Despite shifts in COX and NOS, ß-AR vasodilation is preserved in young adults with obesity. These data highlight the presence of a compensatory shift in microvascular control mechanisms in younger humans with obesity.NEW & NOTEWORTHY We examined ß-adrenergic receptor-mediated vasodilation in skeletal muscle of humans with obesity and normal weight. Results show that despite shifts in the contribution of cyclooxygenase and nitric oxide synthase, ß-adrenergic-mediated vasodilation is relatively preserved in young, otherwise healthy adults with obesity. These data highlight the presence of subclinical changes in microvascular control mechanisms early in the obesity process and suggest duration of obesity and/or the addition of primary aging may be necessary for overt dysfunction.


Assuntos
Músculo Esquelético/irrigação sanguínea , Óxido Nítrico Sintase Tipo III/metabolismo , Obesidade/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Vasodilatação , Agonistas Adrenérgicos beta/farmacologia , Adulto , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Humanos , Isoproterenol/farmacologia , Cetorolaco/farmacologia , Masculino , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/fisiopatologia , Receptores Adrenérgicos beta/metabolismo , ômega-N-Metilarginina/farmacologia
17.
J Mol Med (Berl) ; 100(2): 285-301, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34741638

RESUMO

The risk of severe COVID-19 increases with age as older patients are at highest risk. Thus, there is an urgent need to identify how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) interacts with blood components during aging. We investigated the whole blood transcriptome from the Genotype-Tissue Expression (GTEx) database to explore differentially expressed genes (DEGs) translated into proteins interacting with viral proteins during aging. From 22 DEGs in aged blood, FASLG, CTSW, CTSE, VCAM1, and BAG3 were associated with immune response, inflammation, cell component and adhesion, and platelet activation/aggregation. Males and females older than 50 years old overexpress FASLG, possibly inducing a hyperinflammatory cascade. The expression of cathepsins (CTSW and CTSE) and the anti-apoptotic co-chaperone molecule BAG3 also increased throughout aging in both genders. By exploring single-cell RNA-sequencing data from peripheral blood of SARS-CoV-2-infected patients, we found FASLG and CTSW expressed in natural killer cells and CD8 + T lymphocytes, whereas BAG3 was expressed mainly in CD4 + T cells, naive T cells, and CD14 + monocytes. In addition, T cell exhaustion was associated with increased expression of CCL4L2 and DUSP4 over blood aging. LAG3, PDCD1, TIGIT, VCAM1, HLA-DRA, and TOX also increased in individuals aged 60-69 years old; conversely, the RGS2 gene decreased with aging. We further identified a distinct gene expression profile associated with type I interferon signaling following blood aging. These results revealed changes in blood molecules potentially related to SARS-CoV-2 infection throughout aging, emphasizing them as therapeutic candidates for aggressive clinical manifestation of COVID-19. KEY MESSAGES: • Prediction of host-viral interactions in the whole blood transcriptome during aging. • Expression levels of FASLG, CTSW, CTSE, VCAM1, and BAG3 increase in aged blood. • Blood interactome reveals targets involved with immune response, inflammation, and blood clots. • SARS-CoV-2-infected patients with high viral load showed FASLG overexpression. • Gene expression profile associated with T cell exhaustion and type I interferon signaling were affected with blood aging.


Assuntos
Envelhecimento/sangue , Proteínas Sanguíneas/análise , COVID-19/genética , SARS-CoV-2/patogenicidade , Transcriptoma , Adulto , Idoso , Envelhecimento/genética , Sangue/metabolismo , Análise Química do Sangue , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/virologia , COVID-19/sangue , COVID-19/imunologia , COVID-19/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares/genética , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/virologia , Estudos de Coortes , Feminino , Estudos de Associação Genética , Humanos , Imunidade Inata/genética , Masculino , Pessoa de Meia-Idade , Adulto Jovem
18.
Cardiovasc Res ; 118(1): 97-114, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33135070

RESUMO

Vasculopathy is a pathological process occurring in the blood vessel wall, which could affect the haemostasis and physiological functions of all the vital tissues/organs and is one of the main underlying causes for a variety of human diseases including cardiovascular diseases. Current pharmacological interventions aiming to either delay or stop progression of vasculopathies are suboptimal, thus searching novel, targeted, risk-reducing therapeutic agents, or vascular grafts with full regenerative potential for patients with vascular abnormalities are urgently needed. Since first reported, pluripotent stem cells (PSCs), particularly human-induced PSCs, have open new avenue in all research disciplines including cardiovascular regenerative medicine and disease remodelling. Assisting with recent technological breakthroughs in tissue engineering, in vitro construction of tissue organoid made a tremendous stride in the past decade. In this review, we provide an update of the main signal pathways involved in vascular cell differentiation from human PSCs and an extensive overview of PSC-derived tissue organoids, highlighting the most recent discoveries in the field of blood vessel organoids as well as vascularization of other complex tissue organoids, with the aim of discussing the key cellular and molecular players in generating vascular organoids.


Assuntos
Vasos Sanguíneos/metabolismo , Diferenciação Celular , Linhagem da Célula , Células-Tronco Pluripotentes Induzidas/metabolismo , Neovascularização Fisiológica , Doenças Vasculares/metabolismo , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Técnicas de Cultura de Células , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neovascularização Patológica , Organoides , Fenótipo , Transdução de Sinais , Doenças Vasculares/patologia , Doenças Vasculares/fisiopatologia
19.
Hepatology ; 75(2): 353-368, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34490644

RESUMO

BACKGROUND AND AIMS: Ductular reaction (DR) expands in chronic liver diseases and correlates with disease severity. Besides its potential role in liver regeneration, DR plays a role in the wound-healing response of the liver, promoting periductular fibrosis and inflammatory cell recruitment. However, there is no information regarding its role in intrahepatic angiogenesis. In the current study we investigated the potential contribution of DR cells to hepatic vascular remodeling during chronic liver disease. APPROACH AND RESULTS: In mouse models of liver injury, DR cells express genes involved in angiogenesis. Among angiogenesis-related genes, the expression of Slit2 and its receptor Roundabout 1 (Robo1) was localized in DR cells and neoangiogenic vessels, respectively. The angiogenic role of the Slit2-Robo1 pathway in chronic liver disease was confirmed in ROBO1/2-/+ mice treated with 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which displayed reduced intrahepatic neovascular density compared to wild-type mice. However, ROBO1/2 deficiency did not affect angiogenesis in partial hepatectomy. In patients with advanced alcohol-associated disease, angiogenesis was associated with DR, and up-regulation of SLIT2-ROBO1 correlated with DR and disease severity. In vitro, human liver-derived organoids produced SLIT2 and induced tube formation of endothelial cells. CONCLUSIONS: Overall, our data indicate that DR expansion promotes angiogenesis through the Slit2-Robo1 pathway and recognize DR cells as key players in the liver wound-healing response.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/genética , Hepatopatias Alcoólicas/fisiopatologia , Fígado/fisiopatologia , Neovascularização Patológica/genética , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Animais , Vasos Sanguíneos/metabolismo , Doença Crônica , Progressão da Doença , Expressão Gênica , Ontologia Genética , Hepatite Alcoólica/patologia , Hepatite Alcoólica/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fígado/metabolismo , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/patologia , Camundongos , Neovascularização Patológica/patologia , Neovascularização Fisiológica/genética , Proteínas do Tecido Nervoso/metabolismo , Organoides , Gravidade do Paciente , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Células-Tronco , Regulação para Cima , Remodelação Vascular , Cicatrização
20.
Microvasc Res ; 139: 104262, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34656560

RESUMO

Nanoparticle-based drug targeting is an important platform for the treatment of cardiovascular disorders. Magnetic drug targeting is more significant as it is a noninvasive procedure and biocompatible. The present problem aims to understand magnetic drug delivery to a specific location in a permeable blood vessel under the vibration and magnetic environment. Caputo-Fabrizio fractional-order time derivatives are used in the governing equations. The momentum equations are solved analytically and presented in the form of Lorenzo-Hartley and Robotonov-Hartley functions and convolution of the Laplace transform. Convolution integrations are solved by using the numerical integration technique. The Fourth order Runge-Kutta method (RK4) is used to solve the force balance equation. The influence of pertinent parameters such as Reynolds number, pulsatile frequency, magnetic field strength, Darcy number and fractional-order parameters are presented through graphs. It is observed that increasing Reynolds number results in decreasing the tendency of the drug to capture near the tumor site, whereas the pulsatile frequency presents an opposite phenomenon. Increasing the magnetic field strength and Darcy number boosts the capture efficiency of drug particles near the tumor site. The short memory effect efficiently captures the magnetic drug carriers to a specific location under the action of suitable magnetic field strength.


Assuntos
Antineoplásicos/sangue , Vasos Sanguíneos/metabolismo , Portadores de Fármacos , Campos Magnéticos , Nanopartículas Metálicas , Modelos Cardiovasculares , Neoplasias/irrigação sanguínea , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Simulação por Computador , Composição de Medicamentos , Nanotecnologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Análise Numérica Assistida por Computador , Permeabilidade , Fluxo Sanguíneo Regional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...