Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Physiol Genomics ; 52(8): 314-321, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32628083

RESUMO

Lipocalin 2 (Lcn2) is a multifunctional innate immune protein that limits microbial overgrowth. Our previous study demonstrated that the gut microbiota directly induces intestinal Lcn2 production, and Lcn2-deficient (Lcn2-/-) mice exhibit gut dysbiosis. Coincidentally, gut dysbiosis is associated with metabolic syndrome pathogenesis, and elevated Lcn2 levels has been considered a potential clinical biomarker of metabolic syndrome. Yet whether Lcn2 mitigates or exacerbates metabolic syndrome remains inconclusive. Our objective was to determine whether Lcn2 deficiency-induced compositional changes in gut microbiota contribute to gain in adiposity in aged mice. Utilizing Lcn2-/- mice and their wild-type (WT) littermates, we measured metabolic markers, including fasting blood glucose, serum lipids, fat pad weight, and insulin resistance at ages 3, 6, and 9 mo old. Relative to WT mice, aged Lcn2-/- mice exhibited a gain in adiposity associated with numerous features of metabolic syndrome, including insulin resistance and dyslipidemia. Surprisingly, supplementation with a high-fat diet did not further aggravate metabolic syndrome that spontaneously occurs in Lcn2-/- mice by 6 mo of age. Interestingly, chow-fed Lcn2-/- mice displayed marked differences in the bacterial abundance and metabolomic profile of the gut microbiota compared with WT mice. Overall, our results demonstrate that Lcn2 is essential to maintain metabolic and gut microbiotal homeostasis, where deficiency induces spontaneous delayed onset of metabolic syndrome.


Assuntos
Envelhecimento/metabolismo , Disbiose/complicações , Disbiose/metabolismo , Dislipidemias/complicações , Microbioma Gastrointestinal/genética , Lipocalina-2/deficiência , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Tecido Adiposo , Adiposidade/genética , Animais , Glicemia/análise , Dieta Hiperlipídica , Modelos Animais de Doenças , Disbiose/sangue , Disbiose/microbiologia , Dislipidemias/sangue , Dislipidemias/microbiologia , Feminino , Microbioma Gastrointestinal/imunologia , Homeostase/genética , Lipocalina-2/genética , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo
2.
Physiology (Bethesda) ; 32(3): 224-233, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28404738

RESUMO

Hypertension, or elevated blood pressure (BP), has been extensively researched over decades and clearly demonstrated to be caused due to a combination of host genetic and environmental factors. Although much research remains to be conducted to pin-point the precise genetic elements on the host genome that control BP, new lines of evidence are emerging to indicate that, besides the host genome, the genomes of all indigenous commensal micro-organisms, collectively referred to as the microbial metagenome or microbiome, are important, but largely understudied, determinants of BP. Unlike the rigid host genome, the microbiome or the "second genome" can be altered by diet or microbiotal transplantation in the host. This possibility is attractive from the perspective of exploiting the microbiotal composition for clinical management of inherited hypertension. Thus, focusing on the limited current literature supporting a role for the microbiome in BP regulation, this review highlights the need to further explore the role of the co-existence of host and the microbiota as an organized biological unit called the "holobiont" in the context of BP regulation.


Assuntos
Hipertensão/genética , Hipertensão/microbiologia , Metagenoma , Microbiota , Animais , Humanos , Camundongos
4.
Dis Model Mech ; 13(5)2020 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-32238420

RESUMO

Red blood cell distribution width (RDW) is a measurement of the variation in size and volume of red blood cells (RBCs). Increased RDW, indicating a high heterogeneity of RBCs, is prominently associated with a variety of illnesses, especially cardiovascular diseases. However, the significance of this association to the onset and progression of cardiovascular and renal diseases is unknown. We hypothesized that a genetic predisposition for increased RDW is an early risk factor for cardiovascular and renal comorbidities. Since there is no known animal model of increased RDW, we examined a CRISPR/Cas9 gene-edited rat model (RfflTD) that presented with features of hematologic abnormalities as well as severe cardiac and renal comorbidities. A mass spectrometry-based quantitative proteomic analysis indicated anemia of these rats, which presented with significant downregulation of hemoglobin and haptoglobin. Decreased hemoglobin and increased RDW were further observed in RfflTD through complete blood count. Next, a systematic temporal assessment detected an early increased RDW in RfflTD, which was prior to the development of other comorbidities. The primary mutation of RfflTD is a 50 bp deletion in a non-coding region, and our study has serendipitously identified this locus as a novel quantitative trait locus (QTL) for RDW. To our knowledge, our study is the first to experimentally pinpoint a QTL for RDW and provides a novel genetic rat model mimicking the clinical association of increased RDW with poor cardio-renal outcome.


Assuntos
Doenças Cardiovasculares/genética , Índices de Eritrócitos/genética , Predisposição Genética para Doença , Nefropatias/genética , Animais , Pressão Sanguínea , Peso Corporal , Doenças Cardiovasculares/fisiopatologia , Progressão da Doença , Regulação da Expressão Gênica , Frequência Cardíaca , Doenças Hematológicas/genética , Doenças Hematológicas/fisiopatologia , Hipertrofia , Rim/patologia , Nefropatias/fisiopatologia , Miocárdio/patologia , Condicionamento Físico Animal , Proteômica , Ratos , Fatores de Risco
5.
J Am Heart Assoc ; 9(2): e014373, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31928175

RESUMO

Background Pediatric hypertension is recognized as an emerging global health concern. Although new guidelines are developed for facilitating clinical management, the reasons for the prevalence of hypertension in children remain unknown. Genetics and environmental factors do not fully account for the growing incidence of pediatric hypertension. Because stable bacterial flora in early life are linked with health outcomes later in life, we hypothesized that reshaping of gut microbiota in early life affects blood pressure (BP) of pediatric subjects. Methods and Results To test this hypothesis, we administered amoxicillin, the most commonly prescribed pediatric antibiotic, to alter gut microbiota of young, genetically hypertensive rats (study 1) and dams during gestation and lactation (study 2) and recorded their BP. Reshaping of microbiota with reductions in Firmicutes/Bacteriodetes ratio were observed. Amoxicillin treated rats had lower BP compared with untreated rats. In young rats treated with amoxicillin, the lowering effect on BP persisted even after antibiotics were discontinued. Similarly, offspring from dams treated with amoxicillin showed lower systolic BP compared with control rats. Remarkably, in all cases, a decrease in BP was associated with lowering of Veillonellaceae, which are succinate-producing bacteria. Elevated plasma succinate is reported in hypertension. Accordingly, serum succinate was measured and found lower in animals treated with amoxicillin. Conclusions Our results demonstrate a direct correlation between succinate-producing gut microbiota and early development of hypertension and indicate that reshaping gut microbiota, especially by depleting succinate-producing microbiota early in life, may have long-term benefits for hypertension-prone individuals.


Assuntos
Amoxicilina/farmacologia , Antibacterianos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/prevenção & controle , Veillonellaceae/efeitos dos fármacos , Fatores Etários , Animais , Modelos Animais de Doenças , Feminino , Idade Gestacional , Hipertensão/genética , Hipertensão/microbiologia , Hipertensão/fisiopatologia , Lactação , Masculino , Exposição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos Endogâmicos Dahl , Ácido Succínico/sangue , Veillonellaceae/metabolismo
6.
Hypertension ; 76(1): 59-72, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32450738

RESUMO

Alterations of diurnal rhythms of blood pressure (BP) and reshaping of gut microbiota are both independently associated with hypertension. However, the relationships between biorhythms of BP and gut microbial composition are unknown. We hypothesized that diurnal timing-associated alterations of microbial compositions are synchronous with diurnal rhythmicity, dip in BP, and renal function. To test this hypothesis, Dahl salt-sensitive (S) rats on low- and high-salt diets were examined for time of day effects on gut microbiota, BP, and indicators of renal damage. Major shifts in night and day patterns of specific groups of microbiota were observed between the dark (active) and light (rest) phases, which correlated with diurnal rhythmicity of BP. The diurnal abundance of Firmicutes, Bacteroidetes, and Actinobacteria were independently associated with BP. Discrete bacterial taxa were observed to correlate independently or interactively with one or more of the following 3 factors: (1) BP rhythm, (2) dietary salt, and (3) dip in BP. Phylogenetic Investigation of Communities revealed diurnal timing effects on microbial pathways, characterized by upregulated biosynthetic processes during the active phase of host, and upregulated degradation pathways of metabolites in the resting phase. Additional metagenomics functional pathways with rhythm variations were noted for aromatic amino acid metabolism and taurine metabolism. These diurnal timing dependent changes in microbiota, their functional pathways, and BP dip were associated with concerted effects of the levels of renal lipocalin 2 and kidney injury molecule-1 expression. These data provide evidence for a firm and concerted diurnal timing effects of BP, renal damage, and select microbial communities.


Assuntos
Pressão Sanguínea/fisiologia , Ritmo Circadiano/fisiologia , Microbioma Gastrointestinal/fisiologia , Hipertensão/microbiologia , Rim/efeitos dos fármacos , Cloreto de Sódio na Dieta/administração & dosagem , Ácido 3-Hidroxibutírico/sangue , Animais , Sequência de Bases , Pressão Sanguínea/efeitos dos fármacos , Dieta Hipossódica , Metabolismo Energético , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Genes Bacterianos , Hipertensão/etiologia , Hipertensão/fisiopatologia , Masculino , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Ratos , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta/efeitos adversos
7.
Hypertension ; 72(5): 1125-1132, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30354811

RESUMO

G-protein-coupled estrogen receptor, Gper1, has been implicated in cardiovascular disease, but its mechanistic role in blood pressure control is poorly understood. Here, we demonstrate that genetically salt-sensitive hypertensive rats with complete genomic excision of Gper1 by a multiplexed guide RNA CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 (CRISPR associated proteins) approach present with lower blood pressure, which was accompanied by altered microbiota, different levels of circulating short chain fatty acids, and improved vascular relaxation. Microbiotal transplantation from hypertensive Gper1+/+ rats reversed the cardiovascular protective effect exerted by the genomic deletion of Gper1. Thus, this study reveals a role for Gper1 in promoting microbiotal alterations that contribute to cardiovascular pathology. However, the exact mechanism by which Gper1 regulates blood pressure is still unknown. Our results indicate that the function of Gper1 is contextually dependent on the microbiome, whereby, contemplation of using Gper1 as a target for therapy of cardiovascular disease requires caution.


Assuntos
Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Hipertensão/fisiopatologia , Receptores Acoplados a Proteínas G/genética , Animais , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , RNA Guia de Cinetoplastídeos , Ratos , Ratos Endogâmicos Dahl
8.
Cell Rep ; 25(3): 677-689.e4, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332647

RESUMO

Dietary salt reduction and exercise are lifestyle modifications for salt-sensitive hypertensives. While exercise has prominent metabolic effects, salt has an adverse effect on metabolic syndrome, of which hypertension is a hallmark. We hypothesized that dietary salt impacts metabolism in a salt-sensitive model of hypertension. An untargeted metabolomic approach demonstrates lower circulating levels of the ketone body, beta-hydroxybutyrate (ßOHB), in high salt-fed hypertensive rats. Despite the high salt intake, specific rescue of ßOHB levels by nutritional supplementation of its precursor, 1,3-butanediol, attenuates hypertension and protects kidney function. This beneficial effect of ßOHB was likely independent of gut-microbiotal and Th17-mediated effects of salt and instead facilitated by ßOHB inhibiting the renal Nlrp3 inflammasome. The juxtaposed effects of dietary salt and exercise on salt-sensitive hypertension, which decrease and increase ßOHB respectively, indicate that nutritional supplementation of a precursor of ßOHB provides a similar benefit to salt-sensitive hypertension as exercise.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/prevenção & controle , Inflamassomos/efeitos dos fármacos , Metaboloma/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Cloreto de Sódio na Dieta/toxicidade , Ácido 3-Hidroxibutírico/administração & dosagem , Animais , Pressão Sanguínea , Aromatizantes/toxicidade , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Masculino , Ratos , Ratos Endogâmicos Dahl
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa