Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Pharmacol Res ; 198: 106997, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37972724

RESUMO

Our objective was to investigate whether short-chain fatty acids (SCFAs), specifically acetate and butyrate, could prevent vascular dysfunction and elevated blood pressure (BP) in mice with systemic lupus erythematosus (SLE) induced by TLR7 activation using imiquimod (IMQ). Treatment with both SCFAs and dietary fibers rich in resistant starch (RS) or inulin-type fructans (ITF) effectively prevented the development of hypertension and cardiac hypertrophy. Additionally, these treatments improved aortic relaxation induced by acetylcholine and mitigated vascular oxidative stress. Acetate and butyrate treatments also contributed to the maintenance of colonic integrity, reduced endotoxemia, and decreased the proportion of helper T (Th)17 cells in mesenteric lymph nodes (MLNs), blood, and aorta in TLR7-induced SLE mice. The observed changes in MLNs were correlated with increased levels of GPR43 mRNA in mice treated with acetate and increased GPR41 levels along with decreased histone deacetylase (HDAC)- 3 levels in mice treated with butyrate. Notably, the effects attributed to acetate, but not butyrate, were nullified when co-administered with the GPR43 antagonist GLPG-0974. T cell priming and differentiation into Th17 cells in MLNs, as well as increased Th17 cell infiltration, were linked to aortic endothelial dysfunction and hypertension subsequent to the transfer of faecal microbiota from IMQ-treated mice to germ-free (GF) mice. These effects were counteracted in GF mice through treatment with either acetate or butyrate. To conclude, these findings underscore the potential of SCFA consumption in averting hypertension by restoring balance to the interplay between the gut, immune system, and vascular wall in SLE induced by TLR7 activation.


Assuntos
Microbioma Gastrointestinal , Hipertensão , Lúpus Eritematoso Sistêmico , Microbiota , Animais , Camundongos , Acetatos , Butiratos , Ácidos Graxos Voláteis , Microbioma Gastrointestinal/fisiologia , Hipertensão/prevenção & controle , Receptor 7 Toll-Like
2.
FASEB J ; 34(10): 13626-13640, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32780919

RESUMO

Many probiotics that affect gut microbial ecology have been shown to produce beneficial effects on renin-angiotensin-dependent rodent models and human hypertension. We hypothesized that Bifidobacterium breve CECT7263 (BFM) would attenuate hypertension in deoxycorticosterone acetate (DOCA)-salt rats, a renin-independent model of hypertension. Rats were randomly divided into five groups: control, DOCA-salt, treated DOCA-salt-BFM, treated DOCA-salt-butyrate, and treated DOCA-salt-acetate, for 5 weeks. BFM prevented the increase in systolic blood pressure, cardiac weight, and renal damage induced by DOCA-salt. BFM increased acetate-producing bacterial population and gut acetate levels, improved colonic integrity, normalized endotoxemia, plasma trimethylamine (TMA) levels, and restored the Th17 and Treg content in mesenteric lymph nodes and aorta. Furthermore, BFM improved nitric oxide-dependent vasorelaxation induced by acetylcholine in aortic rings and reduced NADPH oxidase activity in DOCA-salt animals. These protective effects were mimicked by acetate, but not by butyrate supplementation. These data demonstrate that BFM induces changes in gut microbiota linked with attenuation of endothelial dysfunction and increase in blood pressure in this low-renin form of hypertension. These beneficial effects seem to be mediated by increased acetate and reduced TMA production by gut microbiota, thus, improving gut integrity and restoring Th17/Tregs polarization and endotoxemia.


Assuntos
Bifidobacterium breve , Pressão Sanguínea , Microbioma Gastrointestinal , Hipertensão/terapia , Probióticos/uso terapêutico , Vasodilatação , Animais , Acetato de Desoxicorticosterona , Hipertensão/induzido quimicamente , Masculino , Ratos , Ratos Wistar
3.
FASEB J ; 33(9): 10005-10018, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31173526

RESUMO

The aim of the present study was to examine whether the immune-modulatory bacteria Lactobacillus fermentum CECT5716 (LC40) ameliorates disease activity and cardiovascular complications in a female mouse model of lupus. Eighteen-week-old NZBWF1 [systemic lupus erythematosus (SLE)] and NZW/LacJ (control) mice were treated with vehicle or LC40 (5 × 108 colony-forming units/d) for 15 wk. LC40 treatment reduced lupus disease activity, blood pressure, cardiac and renal hypertrophy, and splenomegaly in SLE mice. LC40 reduced the elevated T, B, regulatory T cells (Treg), and T helper (Th)-1 cells in mesenteric lymph nodes of lupus mice. LC40 lowered the higher plasma concentration of proinflammatory cytokines observed in lupus mice. Aortas from SLE mice showed reduced endothelium-dependent vasodilator responses to acetylcholine. Endothelial dysfunction found in SLE is related to an increase of both NADPH oxidase-driven superoxide production and eNOS phosphorylation at the inhibitory Thr495. These activities returned to normal values after a treatment with LC40. Probiotic administration to SLE mice reduced plasma LPS levels, which might be related to an improvement of the gut barrier integrity. LC40 treatment increases the Bifidobacterium count in gut microbiota of SLE mice. In conclusion, our findings identify the gut microbiota manipulation with LC40 as an alternative approach to the prevention of SLE and its associated vascular damage.-Toral, M., Robles-Vera, I., Romero, M., de la Visitación, N., Sánchez, M., O'Valle, F., Rodriguez-Nogales, A., Gálvez, J., Duarte, J., Jiménez, R. Lactobacillus fermentum CECT5716: a novel alternative for the prevention of vascular disorders in a mouse model of systemic lupus erythematosus.


Assuntos
Disbiose/terapia , Limosilactobacillus fermentum , Lúpus Eritematoso Sistêmico/terapia , Probióticos , Doenças Vasculares/prevenção & controle , Acetilcolina/farmacologia , Animais , Aorta/efeitos dos fármacos , Translocação Bacteriana , Bifidobacterium/isolamento & purificação , Citocinas/sangue , Modelos Animais de Doenças , Disbiose/etiologia , Disbiose/microbiologia , Endotoxemia/etiologia , Endotoxemia/prevenção & controle , Feminino , Microbioma Gastrointestinal , Hipertensão/etiologia , Hipertensão/prevenção & controle , Rim/patologia , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/etiologia , Nefrite Lúpica/prevenção & controle , Camundongos , Camundongos Endogâmicos NZB , Miocárdio/patologia , Tamanho do Órgão , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo , Doenças Vasculares/etiologia
4.
Pharmacol Res ; 150: 104487, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31610229

RESUMO

INTRODUCTION: Many studies have showed the beneficial effects of the olive (Olea europaea) leaf extract (OLE) in experimental models of metabolic syndrome, which have been ascribed to the presence of phenolic compounds, like oleuropeoside. This study evaluated the effects of a chemically characterized OLE in high fat diet (HFD)-induced obesity in mice, describing the underlying mechanisms involved in the beneficial effects, with special attention to vascular dysfunction and gut microbiota composition. METHODS: C57BL/6J mice were distributed in different groups: control, control-treated, obese and obese-treated with OLE (1, 10 and 25 mg/kg/day). Control mice received a standard diet, whereas obese mice were fed HFD. The treatment was followed for 5 weeks, and animal body weight periodically assessed. At the end of the treatment, metabolic plasma analysis (including lipid profile) as well as glucose and insulin levels were performed. The HFD-induced inflammatory status was studied in liver and fat, by determining the RNA expression of different inflammatory mediators by qPCR; also, different markers of intestinal epithelial barrier function were determined in colonic tissue by qPCR. Additionally, flow cytometry of immune cells from adipose tissue, endothelial dysfunction in aortic rings as well as gut microbiota composition were evaluated. Faecal microbiota transplantation (FMT) to antibiotic-treated mice fed with HFD was performed. RESULTS: OLE administration reduced body weight gain, basal glycaemia and insulin resistance, and showed improvement in plasma lipid profile when compared with HFD-fed mice. The extract significantly ameliorated the HFD-induced altered expression of key adipogenic genes, like PPARs, adiponectin and leptin receptor, in adipose tissue. Furthermore, the extract reduced the RNA expression of Tnf-α, Il-1ß, Il-6 in liver and adipose tissue, thus improving the tissue inflammatory status associated to obesity. The flow cytometry analysis in adipose tissue corroborated these observations. Additionally, the characterization of the colonic microbiota by sequencing showed that OLE administration was able to counteract the dysbiosis associated to obesity. The extract reversed the endothelial dysfunction observed in the aortic rings of obese mice. FMT from donors HFD-OLE to recipient mice fed an HFD prevented the development of obesity, glucose intolerance, insulin resistance and endothelial dysfunction. CONCLUSION: OLE exerts beneficial effects in HFD-induced obesity in mice, which was associated to an improvement in plasma and tissue metabolic profile, inflammatory status, gut microbiota composition and vascular dysfunction.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Disbiose/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Obesidade/tratamento farmacológico , Olea , Extratos Vegetais/uso terapêutico , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Fármacos Antiobesidade/farmacologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Citocinas/genética , Dieta Hiperlipídica , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Resistência à Insulina , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , Obesidade/metabolismo , Obesidade/microbiologia , Fitoterapia , Extratos Vegetais/farmacologia , Folhas de Planta
5.
Am J Physiol Heart Circ Physiol ; 312(2): H189-H200, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27881385

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARß/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARß/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARß/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARß/δ activators, including molecular and functional mechanisms. Pharmacological PPARß/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARß/δ ligands may provide a basis for the development of therapies in combating hypertension.


Assuntos
Hipertensão/fisiopatologia , PPAR delta/fisiologia , PPAR beta/fisiologia , Vasodilatação/fisiologia , Animais , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Endotélio Vascular/fisiopatologia , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Humanos , Hipertensão/tratamento farmacológico , Inflamação , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR beta/agonistas , PPAR beta/metabolismo , Fenoxiacetatos/farmacologia , Fenoxiacetatos/uso terapêutico , Proteínas RGS/efeitos dos fármacos , Proteínas RGS/genética , Ratos , Ratos Endogâmicos SHR , Sistema Nervoso Simpático/fisiopatologia , Tiazóis/farmacologia , Tiazóis/uso terapêutico , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos
6.
Curr Hypertens Rep ; 19(4): 26, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28315049

RESUMO

PURPOSE OF REVIEW: The present review focuses in the hypertension-associated changes in the microbiota and the current insights regarding the impact of probiotics on blood pressure in animal models and in human hypertensive patients. RECENT FINDINGS: Gut dysbiosis in hypertension is characterized by (i) the gut microbioma that is less diverse and less rich with an increased Firmicutes/Bacteroidetes ratio and (ii) a decrease in acetate- and butyrate-producing bacteria and an increase in lactate-producing bacterial populations. The meta-analysis of the human studies supports that supplementation with probiotics reduces blood pressure. The mechanism of this antihypertensive effect of probiotics and its protective effect on endothelial function has not been fully elucidated. Further investigations are needed to clarify if the effects of probiotic bacteria result from the changes in the gut microbiota and its metabolic by-products; the restoration of the gut barrier function; and the effects on endotoxemia, inflammation, and renal sympathetic nerve activity.


Assuntos
Anti-Hipertensivos/metabolismo , Probióticos , Animais , Pressão Sanguínea/fisiologia , Humanos , Hipertensão/metabolismo , Inflamação/metabolismo , Rim/metabolismo
7.
J Pharmacol Exp Ther ; 358(1): 151-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27189971

RESUMO

Activation of peroxisome proliferator-activated receptor-ß/δ (PPARß) lowers blood pressure in genetic and mineralocorticoid-induced hypertension. Regulator of G-protein-coupled receptor signaling 5 (RGS5) protein, which interferes in angiotensin II (AngII) signaling, is a target gene to PPARß The aim of the present study was to examine whether PPARß activation in resistance arteries and brain tissues prevents the elevated blood pressure in AngII-induced hypertension and evaluate the role of RGS5 in this effect. C57BL/6J male mice were divided into five groups (control mice, PPARß agonist [4-[[[2-[3-Fluoro-4-(trifluoromethyl)phenyl]-4-methyl-5-thiazolyl]methyl]thio]-2-methylphenoxy]acetic acid (GW0742)-treated mice AngII-infused mice, GW0742-treated AngII-infused mice, and AngII-infused mice treated with GW0742 plus PPARß antagonist 3-[[[2-Methoxy-4-(phenylamino)phenyl]amino]sulfonyl]-2-thiophenecarboxylic acid methyl ester (GSK0660)) and were followed for 3 weeks. GW0742 prevented the increase in both arterial blood pressure and plasma noradrenaline levels and the higher reduction of blood pressure after ganglionic blockade, whereas it reduced the mesenteric arterial remodeling and the hyper-responsiveness to vasoconstrictors (AngII and endothelin-1) in AngII-infused mice. These effects were accompanied by an inhibition of NADPH oxidase expression and activity in the brain. Gene expression profiling revealed a marked loss of brainstem and vascular RGS5 in AngII-infused mice, which was restored by GW0742. GW0742-induced effects were abolished by GSK0660. Small interfering RNA targeting RGS5 caused augmented contractile response to AngII in resistance mesenteric arteries and blunted the inhibitory effect of GW0742 on this response. In conclusion, GW0742 exerted antihypertensive effects, restoring sympathetic tone and vascular structure and function in AngII-infused mice by PPARß activation in brain and vessels inhibiting AngII signaling as a result of RGS5 upregulation.


Assuntos
Angiotensina II/farmacologia , Anti-Hipertensivos/uso terapêutico , Hipertensão/tratamento farmacológico , PPAR beta/agonistas , Proteínas RGS/metabolismo , Tiazóis/uso terapêutico , Animais , Anti-Hipertensivos/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/patologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/patologia , Artérias Mesentéricas/fisiopatologia , Camundongos Endogâmicos C57BL , Norepinefrina/sangue , PPAR beta/antagonistas & inibidores , Proteínas RGS/genética , Sulfonas/farmacologia , Tiazóis/administração & dosagem , Tiofenos/farmacologia , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
8.
Clin Sci (Lond) ; 129(9): 823-37, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26253087

RESUMO

Fatty acids cause endothelial dysfunction involving increased ROS (reactive oxygen species) and reduced NO (nitric oxide) bioavailability. We show that in MAECs (mouse aortic endothelial cells), the PPARß/δ (peroxisome- proliferator-activated receptor ß/δ) agonist GW0742 prevented the decreased A23187-stimulated NO production, phosphorylation of eNOS (endothelial nitric oxide synthase) at Ser1177 and increased intracellular ROS levels caused by exposure to palmitate in vitro. The impaired endothelium-dependent relaxation to acetylcholine in mouse aorta induced by palmitate was restored by GW0742. In vivo, GW0742 treatment prevented the reduced aortic relaxation, phosphorylation of eNOS at Ser1177, and increased ROS production and NADPH oxidase in mice fed on a high-fat diet. The PPARß/δ antagonist GSK0660 abolished all of these protective effects induced by GW0742. This agonist enhanced the expression of CPT (carnitine palmitoyltransferase)-1. The effects of GW0742 on acetylcholine- induced relaxation in aorta and on NO and ROS production in MAECs exposed to palmitate were abolished by the CPT-1 inhibitor etomoxir or by siRNA targeting CPT-1. GW0742 also inhibited the increase in DAG (diacylglycerol), PKCα/ßII (protein kinase Cα/ßII) activation, and phosphorylation of eNOS at Thr495 induced by palmitate in MAECs, which were abolished by etomoxir. In conclusion, PPARß/δ activation restored the lipid-induced endothelial dysfunction by up-regulation of CPT-1, thus reducing DAG accumulation and the subsequent PKC-mediated ROS production and eNOS inhibition.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Endotélio Vascular/efeitos dos fármacos , Lipídeos/farmacologia , PPAR delta/metabolismo , PPAR beta/metabolismo , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/fisiologia , Western Blotting , Calcimicina/farmacologia , Carnitina O-Palmitoiltransferase/genética , Células Cultivadas , Diglicerídeos/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/fisiopatologia , Ativação Enzimática/efeitos dos fármacos , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , PPAR delta/agonistas , PPAR delta/antagonistas & inibidores , PPAR beta/agonistas , PPAR beta/antagonistas & inibidores , Ácidos Palmíticos/química , Ácidos Palmíticos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Sulfonas/farmacologia , Tiazóis/farmacologia , Tiofenos/farmacologia , Regulação para Cima
9.
Clin Sci (Lond) ; 127(1): 33-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24410749

RESUMO

Obesity is associated with intestine dysbiosis and is characterized by a low-grade inflammatory status, which affects vascular function. In the present study, we evaluated the effects of a probiotic with immunomodulatory properties, Lactobacillus coryniformis CECT5711, in obese mice fed on an HFD (high-fat diet). The probiotic treatment was given for 12 weeks, and it did not affect the weight evolution, although it reduced basal glycaemia and insulin resistance. L. coryniformis administration to HFD-induced obese mice induced marked changes in microbiota composition and reduced the metabolic endotoxaemia as it decreased the LPS (lipopolysaccharide) plasma level, which was associated with a significant improvement in gut barrier disruption. Furthermore, it lowered TNFα (tumour necrosis factor α) expression in liver, improving the inflammatory status, and thus the glucose metabolism. Additionally, the probiotic reversed the endothelial dysfunction observed in obese mice when endothelium- and NO (nitric oxide)-dependent vasodilatation induced by acetylcholine in aortic rings was studied. It also restored the increased vessel superoxide levels observed in obese mice, by reducing NADPH oxidase activity and increasing antioxidant enzymes. Moreover, chronic probiotic administration for 2 weeks also improved endothelial dysfunction and vascular oxidative stress induced by in vivo administration of LPS in control mice fed on a standard chow diet. The results of the present study demonstrate an endothelial-protective effect of L. coryniformis CECT5711 in obese mice by increasing NO bioavailability, suggesting the therapeutic potential of this gut microbiota manipulation to prevent vasculopathy in obesity.


Assuntos
Endotoxemia/prevenção & controle , Inflamação/terapia , Lactobacillus , Obesidade/complicações , Probióticos/uso terapêutico , Animais , Colo/microbiologia , Dieta Hiperlipídica , Endotélio Vascular/fisiopatologia , Endotoxemia/etiologia , Hiperglicemia/terapia , Inflamação/etiologia , Resistência à Insulina/fisiologia , Lipopolissacarídeos/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Microbiota , Obesidade/microbiologia , Obesidade/fisiopatologia , Oxidantes/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Probióticos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Aumento de Peso/fisiologia
10.
Br J Pharmacol ; 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38770714

RESUMO

BACKGROUND AND PURPOSE: This study analyses whether first-line antihypertensive drugs ameliorate the dysbiosis state in hypertension, and to test if this modification contributes to their blood pressure (BP) lowering properties in a genetic model of neurogenic hypertension. EXPERIMENTAL APPROACH: Twenty-week-old male Wistar Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were untreated or treated with captopril, amlodipine or hydrochlorothiazide. A faecal microbiota transplantation (FMT) experiment was also performed by gavage of faecal content from donor SHR-treated groups to SHR recipients for 3 weeks. KEY RESULTS: Faeces from SHR showed gut dysbiosis, characterized by lower acetate- and higher lactate-producing bacteria and lower strict anaerobic bacteria. All three drugs increased the anaerobic bacteria proportion, captopril and amlodipine restored the proportion of acetate-producing bacterial populations to WKY levels, whereas hydrochlorothiazide decreased butyrate-producing bacteria. Captopril and amlodipine decreased gut pathology and permeability and attenuated sympathetic drive in the gut. Both drugs decreased neuroinflammation and oxidative stress in the hypothalamic paraventricular nuclei. Hydrochlorothiazide was unable to reduce neuroinflammation, gut sympathetic tone and gut integrity. FMT from SHR-amlodipine to SHR decreased BP, ameliorated aortic endothelium-dependent relaxation to acetylcholine, lowered NADPH oxidase activity, aortic Th17 infiltration and reduced neuroinflammation, whereas FMT from SHR-hydrochlorothiazide did not have these effects. CONCLUSIONS AND IMPLICATIONS: First-line antihypertensive drugs induced different modifications of gut integrity and gut dysbiosis in SHR, which result in no contribution of microbiota in the BP lowering effects of hydrochlorothiazide, whereas the vasculo-protective effect induced by amlodipine involves gut microbiota reshaping and gut-immune system communication.

11.
EMBO Mol Med ; 16(1): 132-157, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38177536

RESUMO

Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening condition associated with Marfan syndrome (MFS), a disease caused by fibrillin-1 gene mutations. While various conditions causing TAAD exhibit aortic accumulation of the proteoglycans versican (Vcan) and aggrecan (Acan), it is unclear whether these ECM proteins are involved in aortic disease. Here, we find that Vcan, but not Acan, accumulated in Fbn1C1041G/+ aortas, a mouse model of MFS. Vcan haploinsufficiency protected MFS mice against aortic dilation, and its silencing reverted aortic disease by reducing Nos2 protein expression. Our results suggest that Acan is not an essential contributor to MFS aortopathy. We further demonstrate that Vcan triggers Akt activation and that pharmacological Akt pathway inhibition rapidly regresses aortic dilation and Nos2 expression in MFS mice. Analysis of aortic tissue from MFS human patients revealed accumulation of VCAN and elevated pAKT-S473 staining. Together, these findings reveal that Vcan plays a causative role in MFS aortic disease in vivo by inducing Nos2 via Akt activation and identify Akt signaling pathway components as candidate therapeutic targets.


Assuntos
Aneurisma da Aorta Torácica , Doenças da Aorta , Dissecção Aórtica , Azidas , Desoxiglucose , Síndrome de Marfan , Animais , Humanos , Camundongos , Aneurisma da Aorta Torácica/complicações , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Doenças da Aorta/complicações , Desoxiglucose/análogos & derivados , Síndrome de Marfan/complicações , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Versicanas/metabolismo
12.
Biomed Pharmacother ; 158: 114149, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36566524

RESUMO

Microbiota has a crucial role in the host blood pressure (BP) regulation. The present study analyzes whether the mineralocorticoid receptor antagonist spironolactone ameliorates the dysbiosic state in a genetic model of neurogenic hypertension. Twenty-week-old male Wistar Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were randomly allocated into three groups: untreated WKY, untreated SHR, and SHR treated with spironolactone for 5 weeks. Spironolactone restored the Firmicutes/Bacteroidetes proportion, and acetate-producing bacteria populations to WKY levels. Spironolactone reduced the percentage of intestinal aerobic bacteria. The amelioration of gut dysbiosis was linked to a reduction in the gut pathology, an enhanced colonic integrity, a reduced gut permeability and an attenuated sympathetic drive in the gut. Spironolactone was unable to reduce neuroinflammation and oxidative stress in the paraventricular nuclei in the hypothalamus. Spironolactone reduced the higher Th17 cells proportion in mesenteric lymph nodes and Th17 infiltration in aorta, improved aortic endothelial function and reduced systolic BP. This study demonstrates for the first time that spironolactone reduces gut dysbiosis in SHR. This effect could be related to its capability to improve gut integrity and pathology due to reduced sympathetic drive in the gut.


Assuntos
Microbioma Gastrointestinal , Hipertensão , Animais , Masculino , Ratos , Pressão Sanguínea , Disbiose/tratamento farmacológico , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores de Mineralocorticoides , Espironolactona/farmacologia
13.
Gut Microbes ; 15(2): 2247053, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37615336

RESUMO

This study is to investigate whether dietary fiber intake prevents vascular and renal damage in a genetic mouse model of systemic lupus erythematosus (SLE), and the contribution of gut microbiota in the protective effects. Female NZBWF1 (SLE) mice were treated with resistant-starch (RS) or inulin-type fructans (ITF). In addition, inoculation of fecal microbiota from these experimental groups to recipient normotensive female C57Bl/6J germ-free (GF) mice was performed. Both fiber treatments, especially RS, prevented the development of hypertension, renal injury, improved the aortic relaxation induced by acetylcholine, and the vascular oxidative stress. RS and ITF treatments increased the proportion of acetate- and butyrate-producing bacteria, respectively, improved colonic inflammation and integrity, endotoxemia, and decreased helper T (Th)17 proportion in mesenteric lymph nodes (MLNs), blood, and aorta in SLE mice. However, disease activity (splenomegaly and anti-ds-DNA) was unaffected by both fibers. T cell priming and Th17 differentiation in MLNs and increased Th17 infiltration was linked to aortic endothelial dysfunction and hypertension after inoculation of fecal microbiota from SLE mice to GF mice, without changes in proteinuria and autoimmunity. All these effects were lower in GF mice after fecal inoculation from fiber-treated SLE mice. In conclusion, these findings support that fiber consumption prevented the development of hypertension by rebalancing of dysfunctional gut-immune system-vascular wall axis in SLE.


Assuntos
Microbioma Gastrointestinal , Hipertensão , Lúpus Eritematoso Sistêmico , Microbiota , Feminino , Animais , Camundongos , Fibras na Dieta , Amido Resistente , Lúpus Eritematoso Sistêmico/complicações
14.
Br J Pharmacol ; 179(7): 1287-1303, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34599830

RESUMO

Recent studies have shown that NO is a central mediator in diseases associated with thoracic aortic aneurysm, such as Marfan syndrome. The progressive dilation of the aorta in thoracic aortic aneurysm ultimately leads to aortic dissection. Unfortunately, current medical treatments have neither halt aortic enlargement nor prevented rupture, leaving surgical repair as the only effective treatment. There is therefore a pressing need for effective therapies to delay or even avoid the need for surgical repair in thoracic aortic aneurysm patients. Here, we summarize the mechanisms through which NO signalling dysregulation causes thoracic aortic aneurysm, particularly in Marfan syndrome. We discuss recent advances based on the identification of new Marfan syndrome mediators related to pathway overactivation that represent potential disease biomarkers. Likewise, we propose iNOS, sGC and PRKG1, whose pharmacological inhibition reverses aortopathy in Marfan syndrome mice, as targets for therapeutic intervention in thoracic aortic aneurysm and are candidates for clinical trials.


Assuntos
Aneurisma da Aorta Torácica , Aneurisma Aórtico , Dissecção Aórtica , Síndrome de Marfan , Dissecção Aórtica/complicações , Dissecção Aórtica/cirurgia , Animais , Aorta , Aneurisma Aórtico/complicações , Aneurisma da Aorta Torácica/etiologia , Aneurisma da Aorta Torácica/prevenção & controle , Aneurisma da Aorta Torácica/cirurgia , Proteína Quinase Dependente de GMP Cíclico Tipo I , Humanos , Síndrome de Marfan/complicações , Síndrome de Marfan/tratamento farmacológico , Síndrome de Marfan/cirurgia , Camundongos
15.
Biomed Pharmacother ; 153: 113445, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36076560

RESUMO

BACKGROUND AND PURPOSE: Melatonin has shown beneficial effects on obesity, both in humans and experimental models, via regulating the altered circadian rhythm and thus ameliorating the gut dysbiosis associated with this metabolic condition. However, its clinical use is limited, mostly due to its short half-life. Agomelatine is an agonist of the melatonin receptors that could be used to manage obesity and offer a better profile than melatonin. EXPERIMENTAL APPROACH: Male C57BL/6 mice were fed a high fat diet and orally treated for five weeks with agomelatine, or melatonin or metformin, used as control drugs. Metabolic profile, inflammatory status, vascular dysfunction and intestinal microbiota composition were assessed. KEY RESULTS: Agomelatine lessened body weight gain, enhanced glucose and lipid metabolisms, and improved insulin resistance. It also reduced the obesity-associated inflammatory status and endothelial dysfunction, probably linked to its effect on gut dysbiosis, consisting of the restoration of bacterial populations with key functions, such as short chain fatty acid production. CONCLUSIONS AND IMPLICATIONS: Agomelatine can be considered as a novel therapeutic tool for the management of human obesity and its associated comorbidities.


Assuntos
Microbioma Gastrointestinal , Melatonina , Acetamidas , Animais , Dieta Hiperlipídica/efeitos adversos , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Humanos , Masculino , Melatonina/farmacologia , Melatonina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Naftalenos , Obesidade/metabolismo
16.
Biomed Pharmacother ; 135: 111189, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33388596

RESUMO

Microbiota has a role in the host blood pressure (BP) regulation. The immunosuppressive drug mofetil mycophenolate (MMF) ameliorates hypertension. The present study analyzes whether MMF improves dysbiosis in a genetic model of hypertension. Twenty weeks old male spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY) were randomly divided into three groups: untreated WKY, untreated SHR, and SHR treated with MMF for 5 weeks. MMF treatment restored gut bacteria from the phyla Firmicutes and Bacteroidetes, and acetate- and lactate-producing bacteria to levels similar to those found in WKY, increasing butyrate-producing bacteria. MMF increased the percentage of anaerobic bacteria in the gut. The improvement of gut dysbiosis was associated with an enhanced colonic integrity and a decreased sympathetic drive in the gut. MMF inhibited neuroinflammation in the paraventricular nuclei in the hypothalamus. MMF increased the lower regulatory T cells proportion in mesenteric lymph nodes and Th17 and Th1 infiltration in aorta, improved aortic endothelial function and reduced systolic BP. This study demonstrates for the first time that MMF reduces gut dysbiosis in SHR. This effect could be related to its capability to improve gut integrity due to reduced sympathetic drive in the gut associated to the reduced brain neuroinflammation.


Assuntos
Anti-Hipertensivos/farmacologia , Bactérias/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Colo/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Ácido Micofenólico/farmacologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/imunologia , Aorta Torácica/fisiopatologia , Bactérias/crescimento & desenvolvimento , Bactérias/metabolismo , Células Cultivadas , Colo/inervação , Citocinas/metabolismo , Modelos Animais de Doenças , Disbiose , Hipertensão/imunologia , Hipertensão/microbiologia , Hipertensão/fisiopatologia , Mediadores da Inflamação/metabolismo , Neuroimunomodulação/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
17.
Nutrients ; 13(9)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34578849

RESUMO

Previous experiments in animals and humans show that shifts in microbiota and its metabolites are linked to hypertension. The present study investigates whether doxycycline (DOX, a broad-spectrum tetracycline antibiotic) improves dysbiosis, prevent cardiovascular pathology and attenuate hypertension in deoxycorticosterone acetate (DOCA)-salt rats, a renin-independent model of hypertension. Male Wistar rats were randomly assigned to three groups: control, DOCA-salt hypertensive rats, DOCA-salt treated with DOX for 4 weeks. DOX decreased systolic blood pressure, improving endothelial dysfunction and reducing aortic oxidative stress and inflammation. DOX decreased lactate-producing bacterial population and plasma lactate levels, improved gut barrier integrity, normalized endotoxemia, plasma noradrenaline levels and restored the Treg content in aorta. These data demonstrate that DOX through direct effects on gut microbiota and its non-microbial effects (anti-inflammatory and immunomodulatory) reduces endothelial dysfunction and the increase in blood pressure in this low-renin form of hypertension.


Assuntos
Antibacterianos/farmacologia , Doxiciclina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Animais , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Endotélio Vascular/fisiopatologia , Hipertensão/fisiopatologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Vasodilatação/efeitos dos fármacos
18.
Antioxidants (Basel) ; 10(9)2021 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-34573058

RESUMO

Our group has investigated the involvement of gut microbiota in hypertension in a murine model of systemic lupus erythematosus induced by Toll-like receptor (TLR)-7 activation. Female BALB/c mice were randomly assigned to four experimental groups: an untreated control (CTR), a group treated with the TLR7 agonist imiquimod (IMQ), IMQ-treated with vancomycin, and IMQ-treated with a cocktail of broad-spectrum antibiotics. We carried out faecal microbiota transplant (FMT) from donor CTR or IMQ mice to recipient IMQ or CTR animals, respectively. Vancomycin inhibited the increase in blood pressure; improved kidney injury, endothelial function, and oxidative stress; and reduced T helper (Th)17 infiltration in aortas from IMQ-treated mice. The rise in blood pressure and vascular complications present in IMQ mice were also observed in the CTR mice recipients of IMQ microbiota. Reduced relative populations of Sutterella and Anaerovibrio were associated with high blood pressure in our animals, which were increased after stool transplantation of healthy microbiota to IMQ mice. The reduced endothelium-dependent vasodilator responses to acetylcholine induced by IMQ microbiota were normalized after interleukin-17 neutralization. In conclusion, gut microbiota plays a role in the TLR7-driven increase in Th17 cell, endothelial dysfunction, vascular inflammation, and hypertension. The vascular changes induced by IMQ microbiota were initiated by Th17 infiltrating the vasculature.

19.
Nutrients ; 13(8)2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34444829

RESUMO

Our group tested the effects of Lactobacillus fermentum CECT5716 (LC40) and/or Bifidobacterium breve CECT7263 (BFM) in the prevention of gut dysbiosis, hypertension and endothelial dysfunction in a pharmacologically-induced model of systemic lupus erythematosus (SLE). We treated eight-week-old BALB/cByJRj mice without (Ctrl) or with the agonist of TLR-7 Imiquimod (IMQ) for 8 weeks. Concomitantly, LC40 (109 CFU/mL) and BFM (109 CFU/mL) were administered through oral gavage once a day. IMQ induced intestinal dysbiosis consisting of a decrease in the α-diversity measured with Chao-richness and numbers of species. LC40 and BFM did not restore these parameters. The three-dimensional principal component analysis of bacterial taxa in stool samples presented perfect clustering between Ctrl and IMQ groups. Clusters corresponding to LC40 and BFM were more akin to IMQ. BFM and LC40 were detected colonizing the gut microbiota of mice treated respectively. LC40 and BFM decreased plasma double-stranded DNA autoantibodies, and B cells in spleen, which were increased in the IMQ group. Also, LC40 and BFM treatments activated TLR9, reduced T cells activation, and Th17 polarization in mesenteric lymph nodes. Aortae from IMQ mice displayed a decreased endothelium-dependent vasodilator response to acetylcholine linked to pro-inflammatory and pro-oxidative status, which were normalized by both BFM and LC40. In conclusion, we demonstrate for the first time that the chronic treatment with LC40 or BFM prevented hypertension and endothelial dysfunction in a mouse lupus model induced by TLR-7 activation.


Assuntos
Hipertensão/prevenção & controle , Lúpus Eritematoso Sistêmico/complicações , Probióticos/uso terapêutico , Receptor 7 Toll-Like/agonistas , Animais , Bifidobacterium breve , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Disbiose/microbiologia , Feminino , Microbioma Gastrointestinal , Imunidade , Limosilactobacillus fermentum , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Ribossômico 16S , Espécies Reativas de Oxigênio
20.
Br J Pharmacol ; 178(18): 3708-3729, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33931880

RESUMO

BACKGROUND AND PURPOSE: Hypertension is an important cardiovascular risk factor that is prevalent in the systemic lupus erythematosus patient population. Here, we have investigated whether intestinal microbiota is involved in hypertension in a genetic mouse model of systemic lupus erythematosus. EXPERIMENTAL APPROACH: Twenty-six-week-old female NZW/LacJ (control) and NZBWF1 (F1 hybrid of New Zealand Black and New Zealand White strains; systemic lupus erythematosus) mice were treated for 6 weeks with a broad-spectrum antibiotic mixture or with vancomycin. Faecal microbiota transplantation was performed from donor systemic lupus erythematosus group to recipient to germ-depleted or germ-free mice. KEY RESULTS: Antibiotic treatment inhibited the development of hypertension and renal injury, improved endothelial dysfunction and vascular oxidative stress, and decreased aortic Th17 infiltration in NZBWF1 mice. High BP and vascular complications found in systemic lupus erythematosus mice, but not autoimmunity, kidney inflammation and endotoxemia, were reproduced by the transfer of gut microbiota from systemic lupus erythematosus donors to germ-free or germ-depleted mice. Increased proportions of Bacteroides were linked with high BP in these mice. The reduced endothelium-dependent vasodilator responses to acetylcholine and the high BP induced by microbiota from hypertensive systemic lupus erythematosus mice were inhibited after IL-17 neutralization. CONCLUSION AND IMPLICATIONS: Changes in T-cell populations, endothelial function, vascular inflammation and hypertension driven by a genetic systemic lupus erythematosus background can be modified by antibiotic-induced changes in gut microbiota. The vascular changes induced by hypertensive systemic lupus erythematosus microbiota were mediated by Th17 infiltration in the vasculature.


Assuntos
Microbioma Gastrointestinal , Hipertensão , Lúpus Eritematoso Sistêmico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Rim , Camundongos , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA