Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 326(6): F1032-F1038, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38634136

RESUMO

The gut microbiome regulates many important host physiological processes associated with cardiovascular health and disease; however, the impact of the gut microbiome on aldosterone is unclear. Investigating whether gut microbiota regulate aldosterone can offer novel insights into how the microbiome affects blood pressure. In this study, we aimed to determine whether gut microbiota regulate host aldosterone. We used enzyme-linked immunosorbent assays (ELISAs) to assess plasma aldosterone and plasma renin activity (PRA) in female and male mice in which gut microbiota are intact, suppressed, or absent. In addition, we examined urinary aldosterone. Our findings demonstrated that when the gut microbiota is suppressed following antibiotic treatment, there is an increase in plasma and urinary aldosterone in both female and male mice. In contrast, an increase in PRA is seen only in males. We also found that when gut microbiota are absent (germ-free mice), plasma aldosterone is significantly increased compared with conventional animals (in both females and males), but PRA is not. Understanding how gut microbiota influence aldosterone levels could provide valuable insights into the development and treatment of hypertension and/or primary aldosteronism. This knowledge may open new avenues for therapeutic interventions, such as probiotics or dietary modifications to help regulate blood pressure via microbiota-based changes to aldosterone.NEW & NOTEWORTHY We explore the role of the gut microbiome in regulating aldosterone, a hormone closely linked to blood pressure and cardiovascular disease. Despite the recognized importance of the gut microbiome in host physiology, the relationship with circulating aldosterone remains largely unexplored. We demonstrate that suppression of gut microbiota leads to increased levels of plasma and urinary aldosterone. These findings underscore the potential of the gut microbiota to influence aldosterone regulation, suggesting new possibilities for treating hypertension.


Assuntos
Aldosterona , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Renina , Animais , Aldosterona/sangue , Aldosterona/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Feminino , Masculino , Renina/sangue , Renina/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Vida Livre de Germes , Camundongos , Antibacterianos/farmacologia , Hipertensão/microbiologia , Hipertensão/metabolismo
2.
Am J Physiol Renal Physiol ; 324(6): F511-F520, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37053490

RESUMO

The gut microbiome impacts host gene expression not only in the colon but also at distal sites including the liver, white adipose tissue, and spleen. The gut microbiome also influences the kidney and is associated with renal diseases and pathologies; however, a role for the gut microbiome to modulate renal gene expression has not been examined. To determine if microbes modulate renal gene expression, we used whole organ RNA sequencing to compare gene expression in C57Bl/6 mice that were germ free (lacking gut microbiota) versus conventionalized (gut microbiota reintroduced using an oral gavage of a fecal slurry composed of mixed stool). 16S sequencing showed that male and female mice were similarly conventionalized, although Verrucomicrobia was higher in male mice. We found that renal gene expression was differentially regulated in the presence vs. absence of microbiota and that these changes were largely sex specific. Although microbes also influenced gene expression in the liver and large intestine, most differentially expressed genes (DEGs) in the kidney were not similarly regulated in the liver or large intestine. This demonstrates that the influence of the gut microbiota on gene expression is tissue specific. However, a minority of genes (n = 4 in males and n = 6 in females) were similarly regulated in all three tissues examined, including genes associated with circadian rhythm (period 1 in males and period 2 in females) and metal binding (metallothionein 1 and metallothionein 2 in both males and females). Finally, using a previously published single-cell RNA-sequencing dataset, we assigned a subset of DEGs to specific kidney cell types, revealing clustering of DEGs by cell type and/or sex.NEW & NOTEWORTHY It is unknown whether the microbiome influences host gene expression in the kidney. Here, we utilized an unbiased, bulk RNA-sequencing approach to compare gene expression in the kidneys of male and female mice with or without gut microbiota. This report demonstrates that renal gene expression is modulated by the microbiome in a sex- and tissue-specific manner.


Assuntos
Microbioma Gastrointestinal , Masculino , Feminino , Animais , Camundongos , Fezes , Colo , Rim , Expressão Gênica
3.
Am J Physiol Renal Physiol ; 324(3): F267-F273, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36603001

RESUMO

The diverse functions of each nephron segment rely on the coordinated action of specialized cell populations that are uniquely defined by their transcriptional profile. In the collecting duct, there are two critical and distinct cell populations: principal cells and intercalated cells. Principal cells play key roles in the regulation of water, Na+, and K+, whereas intercalated cells are best known for their role in acid-base homeostasis. Currently, there are no in vitro systems that recapitulate the heterogeneity of the collecting ducts, which limits high-throughput and replicate investigations of genetic and physiological phenomena. Here, we demonstrated that the transcription factor Foxi1 is sufficient to alter the transcriptional identity of M-1 cells, a murine cortical collecting duct cell line. Specifically, overexpression of Foxi1 induces the expression of intercalated cell transcripts including Gpr116, Atp6v1b1, Atp6v1g3, Atp6v0d2, Slc4a9, and Slc26a4. These data indicate that overexpression of Foxi1 differentiates M-1 cells toward a non-A, non-B type intercalated cell phenotype and may provide a novel in vitro tool to study transcriptional regulation and physiological function of the renal collecting duct.NEW & NOTEWORTHY Transfection of M-1 cells with the transcription factor Foxi1 generates cells that express V-ATPase and Gpr116 as well as other genes associated with renal intercalated cells. This straightforward and novel in vitro system could be used to study processes including transcriptional regulation and cell specification and differentiation in renal intercalated cells.


Assuntos
Fatores de Transcrição Forkhead , Receptores Acoplados a Proteínas G , ATPases Vacuolares Próton-Translocadoras , Animais , Camundongos , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Diferenciação Celular , Antiportadores de Cloreto-Bicarbonato/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Rim/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(42): 26470-26481, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33004624

RESUMO

The diversity and near universal expression of G protein-coupled receptors (GPCR) reflects their involvement in most physiological processes. The GPCR superfamily is the largest in the human genome, and GPCRs are common pharmaceutical targets. Therefore, uncovering the function of understudied GPCRs provides a wealth of untapped therapeutic potential. We previously identified an adhesion-class GPCR, Gpr116, as one of the most abundant GPCRs in the kidney. Here, we show that Gpr116 is highly expressed in specialized acid-secreting A-intercalated cells (A-ICs) in the kidney using both imaging and functional studies, and we demonstrate in situ receptor activation using a synthetic agonist peptide unique to Gpr116. Kidney-specific knockout (KO) of Gpr116 caused a significant reduction in urine pH (i.e., acidification) accompanied by an increase in blood pH and a decrease in pCO2 compared to WT littermates. Additionally, immunogold electron microscopy shows a greater accumulation of V-ATPase proton pumps at the apical surface of A-ICs in KO mice compared to controls. Furthermore, pretreatment of split-open collecting ducts with the synthetic agonist peptide significantly inhibits proton flux in ICs. These data suggest a tonic inhibitory role for Gpr116 in the regulation of V-ATPase trafficking and urinary acidification. Thus, the absence of Gpr116 results in a primary excretion of acid in KO mouse urine, leading to mild metabolic alkalosis ("renal tubular alkalosis"). In conclusion, we have uncovered a significant role for Gpr116 in kidney physiology, which may further inform studies in other organ systems that express this GPCR, such as the lung, testes, and small intestine.


Assuntos
Rim/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Fenômenos Bioquímicos , Transporte Biológico , Movimento Celular/fisiologia , Células Epiteliais/metabolismo , Feminino , Homeostase , Humanos , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos Knockout
5.
Proc Natl Acad Sci U S A ; 117(45): 28485-28495, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33097666

RESUMO

The recent discovery of sensory (tastant and odorant) G protein-coupled receptors on the smooth muscle of human bronchi suggests unappreciated therapeutic targets in the management of obstructive lung diseases. Here we have characterized the effects of a wide range of volatile odorants on the contractile state of airway smooth muscle (ASM) and uncovered a complex mechanism of odorant-evoked signaling properties that regulate excitation-contraction (E-C) coupling in human ASM cells. Initial studies established multiple odorous molecules capable of increasing intracellular calcium ([Ca2+]i) in ASM cells, some of which were (paradoxically) associated with ASM relaxation. Subsequent studies showed a terpenoid molecule (nerol)-stimulated OR2W3 caused increases in [Ca2+]i and relaxation of ASM cells. Of note, OR2W3-evoked [Ca2+]i mobilization and ASM relaxation required Ca2+ flux through the store-operated calcium entry (SOCE) pathway and accompanied plasma membrane depolarization. This chemosensory odorant receptor response was not mediated by adenylyl cyclase (AC)/cyclic nucleotide-gated (CNG) channels or by protein kinase A (PKA) activity. Instead, ASM olfactory responses to the monoterpene nerol were predominated by the activity of Ca2+-activated chloride channels (TMEM16A), including the cystic fibrosis transmembrane conductance regulator (CFTR) expressed on endo(sarco)plasmic reticulum. These findings demonstrate compartmentalization of Ca2+ signals dictates the odorant receptor OR2W3-induced ASM relaxation and identify a previously unrecognized E-C coupling mechanism that could be exploited in the development of therapeutics to treat obstructive lung diseases.


Assuntos
Anoctamina-1/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Odorantes/metabolismo , Adenilil Ciclases/metabolismo , Brônquios/metabolismo , Cálcio/metabolismo , Células Cultivadas , Humanos , Pulmão/metabolismo , Contração Muscular/fisiologia , Relaxamento Muscular , Miócitos de Músculo Liso/metabolismo , Receptores Odorantes/genética
6.
Am J Physiol Cell Physiol ; 322(6): C1279-C1288, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35544696

RESUMO

In this study, we elucidate factors that regulate the trafficking and activity of a well-conserved olfactory receptor (OR), olfactory receptor 558 (Olfr558), and its human ortholog olfactory receptor 51E1 (OR51E1). Results indicate that butyrate activates Olfr558/OR51E1 leading to the production of cAMP, and evokes Ca2+ influx. We also find olfactory G protein (Golf) increases cAMP production induced by Olfr558/OR51E1 activation but does not affect trafficking. Given the 93% sequence identity between OR51E1 and Olfr558, it is surprising to note that OR51E1 has significantly more surface expression yet similar total protein expression. We find that replacing the Olfr558 N-terminus with that of OR51E1 significantly increases trafficking; in contrast, there is no change in surface expression conferred by the OR51E1 TM2, TM3, or TM4 domains. A previous analysis of human OR51E1 single nucleotide polymorphisms (SNPs) identified an A156T mutant primarily found in South Asia as the most abundant (albeit still rare). We find that the OR51E1 A156T mutant has reduced surface expression and cAMP production without a change in total protein expression. In sum, this study of a well-conserved olfactory receptor identifies both protein regions and specific amino acid residues that play key roles in protein trafficking and also elucidates common effects of Golf on the regulation of both the human and murine OR.


Assuntos
Receptores Odorantes , Aminoácidos/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo , Receptores Odorantes/genética
7.
Physiology (Bethesda) ; 35(4): 275-284, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32490748

RESUMO

Shifts in the gut microbiome play a key role in blood pressure regulation, and changes in the production of gut microbial metabolites are likely to be a key mechanism. Known gut microbial metabolites include short-chain fatty acids, which can signal via G-protein-coupled receptors, and trimethylamine-N oxide. In this review, we provide an overview of gut microbial metabolites documented thus far to play a role in blood pressure regulation.


Assuntos
Ácidos Graxos Voláteis/metabolismo , Hipertensão/microbiologia , Hipertensão/fisiopatologia , Metilaminas/metabolismo , Animais , Pressão Sanguínea/fisiologia , Microbioma Gastrointestinal , Humanos , Hipertensão/metabolismo
8.
J Pharmacol Exp Ther ; 377(1): 39-50, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33414131

RESUMO

Short-chain fatty acids (SCFAs) are metabolites produced almost exclusively by the gut microbiota and are an essential mechanism by which gut microbes influence host physiology. Given that SCFAs induce vasodilation, we hypothesized that they might have additional cardiovascular effects. In this study, novel mechanisms of SCFA action were uncovered by examining the acute effects of SCFAs on cardiovascular physiology in vivo and ex vivo. Acute delivery of SCFAs in conscious radiotelemetry-implanted mice results in a simultaneous decrease in both mean arterial pressure and heart rate (HR). Inhibition of sympathetic tone by the selective ß-1 adrenergic receptor antagonist atenolol blocks the acute drop in HR seen with acetate administration, yet the decrease in mean arterial pressure persists. Treatment with tyramine, an indirect sympathomimetic, also blocks the acetate-induced acute drop in HR. Langendorff preparations show that acetate lowers HR only after long-term exposure and at a smaller magnitude than seen in vivo. Pressure-volume loops after acetate injection show a decrease in load-independent measures of cardiac contractility. Isolated trabecular muscle preparations also show a reduction in force generation upon SCFA treatment, though only at supraphysiological concentrations. These experiments demonstrate a direct cardiac component of the SCFA cardiovascular response. These data show that acetate affects blood pressure and cardiac function through parallel mechanisms and establish a role for SCFAs in modulating sympathetic tone and cardiac contractility, further advancing our understanding of the role of SCFAs in blood pressure regulation. SIGNIFICANCE STATEMENT: Acetate, a short-chain fatty acid, acutely lowers heart rate (HR) as well as mean arterial pressure in vivo in radiotelemetry-implanted mice. Acetate is acting in a sympatholytic manner on HR and exerts negative inotropic effects in vivo. This work has implications for potential short-chain fatty acid therapeutics as well as gut dysbiosis-related disease states.


Assuntos
Acetatos/farmacologia , Pressão Sanguínea , Ácidos Graxos Voláteis/farmacologia , Frequência Cardíaca , Coração/efeitos dos fármacos , Contração Miocárdica , Acetatos/administração & dosagem , Animais , Ácidos Graxos Voláteis/administração & dosagem , Feminino , Coração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiologia
9.
Am J Physiol Regul Integr Comp Physiol ; 320(1): R19-R35, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33085906

RESUMO

C1q/TNF-related protein 1 (CTRP1) is an endocrine factor with metabolic, cardiovascular, and renal functions. We previously showed that aged Ctrp1-knockout (KO) mice fed a control low-fat diet develop renal hypertrophy and dysfunction. Since aging and obesity adversely affect various organ systems, we hypothesized that aging, in combination with obesity induced by chronic high-fat feeding, would further exacerbate renal dysfunction in CTRP1-deficient animals. To test this, we fed wild-type and Ctrp1-KO mice a high-fat diet for 8 mo or longer. Contrary to our expectation, no differences were observed in blood pressure, heart function, or vascular stiffness between genotypes. Loss of CTRP1, however, resulted in an approximately twofold renal enlargement (relative to body weight), ∼60% increase in urinary total protein content, and elevated pH, and changes in renal gene expression affecting metabolism, signaling, transcription, cell adhesion, solute and metabolite transport, and inflammation. Assessment of glomerular integrity, the extent of podocyte foot process effacement, as well as renal response to water restriction and salt loading did not reveal significant differences between genotypes. Interestingly, blood platelet, white blood cell, neutrophil, lymphocyte, and eosinophil counts were significantly elevated, whereas mean corpuscular volume and hemoglobin were reduced in Ctrp1-KO mice. Cytokine profiling revealed increased circulating levels of CCL17 and TIMP-1 in KO mice. Compared with our previous study, current data suggest that chronic high-fat feeding affects renal phenotypes differently than similarly aged mice fed a control low-fat diet, highlighting a diet-dependent contribution of CTRP1 deficiency to age-related changes in renal structure and function.


Assuntos
Adipocinas/deficiência , Envelhecimento/metabolismo , Dieta Hiperlipídica/efeitos adversos , Nefropatias/etiologia , Rim/metabolismo , Obesidade/etiologia , Adipocinas/genética , Fatores Etários , Envelhecimento/genética , Envelhecimento/patologia , Animais , Quimiocina CCL17/sangue , Feminino , Regulação da Expressão Gênica , Genótipo , Hipertrofia , Rim/ultraestrutura , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Inibidor Tecidual de Metaloproteinase-1/sangue
10.
FASEB J ; 34(2): 2657-2676, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908037

RESUMO

Local and systemic factors that influence renal structure and function in aging are not well understood. The secretory protein C1q/TNF-related protein 1 (CTRP1) regulates systemic metabolism and cardiovascular function. We provide evidence here that CTRP1 also modulates renal physiology in an age- and sex-dependent manner. In mice lacking CTRP1, we observed significantly increased kidney weight and glomerular hypertrophy in aged male but not female or young mice. Although glomerular filtration rate, plasma renin and aldosterone levels, and renal response to water restriction did not differ between genotypes, CTRP1-deficient male mice had elevated blood pressure. Echocardiogram and pulse wave velocity measurements indicated normal heart function and vascular stiffness in CTRP1-deficient animals, and increased blood pressure was not due to greater salt retention. Paradoxically, CTRP1-deficient mice had elevated urinary sodium and potassium excretion, partially resulting from reduced expression of genes involved in renal sodium and potassium reabsorption. Despite renal hypertrophy, markers of inflammation, fibrosis, and oxidative stress were reduced in CTRP1-deficient mice. RNA sequencing revealed alterations and enrichments of genes in metabolic processes in CTRP1-deficient animals. These results highlight novel contributions of CTRP1 to aging-associated changes in renal physiology.


Assuntos
Adipocinas/deficiência , Hipertensão/metabolismo , Hipertrofia/metabolismo , Rim/metabolismo , Adipocinas/metabolismo , Animais , Pressão Sanguínea/fisiologia , Hipertensão/fisiopatologia , Hipertrofia/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Camundongos Knockout , Transdução de Sinais/fisiologia
11.
Annu Rev Physiol ; 78: 391-414, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26667077

RESUMO

To maintain metabolic homeostasis, the body must be able to monitor the concentration of a large number of substances, including metabolites, in real time and to use that information to regulate the activities of different metabolic pathways. Such regulation is achieved by the presence of sensors, termed metabolite receptors, in various tissues and cells of the body, which in turn convey the information to appropriate regulatory or positive or negative feedback systems. In this review, we cover the unique roles of metabolite receptors in renal and vascular function. These receptors play a wide variety of important roles in maintaining various aspects of homeostasis-from salt and water balance to metabolism-by sensing metabolites from a wide variety of sources. We discuss the role of metabolite sensors in sensing metabolites generated locally, metabolites generated at distant tissues or organs, or even metabolites generated by resident microbes. Metabolite receptors are also involved in various pathophysiological conditions and are being recognized as potential targets for new drugs. By highlighting three receptor families-(a) citric acid cycle intermediate receptors, (b) purinergic receptors, and


Assuntos
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , Homeostase/fisiologia , Rim/metabolismo , Rim/fisiologia , Receptores de Superfície Celular/metabolismo , Animais , Humanos
12.
Am J Physiol Renal Physiol ; 316(2): F372-F381, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30484350

RESUMO

Olfactory receptors are G protein-coupled receptors that serve to detect odorants in the nose. Additionally, these receptors are expressed in other tissues, where they have functions outside the canonical smell response. Olfactory receptor 1393 (Olfr1393) was recently identified as a novel regulator of Na+-glucose cotransporter 1 (Sglt1) localization in the renal proximal tubule. Glucose reabsorption in the proximal tubule (via Sglt1 and Sglt2) has emerged as an important contributor to the development of diabetes. Inhibition of Sglt2 is accepted as a viable therapeutic treatment option for patients with type 2 diabetes and has been shown to delay development of diabetic kidney disease. We hypothesized that Olfr1393 may contribute to the progression of type 2 diabetes, particularly the development of hyperfiltration, which has been linked to increased Na+ reabsorption in the proximal tubule via the Sglts. To test this hypothesis, Olfr1393 wild-type (WT) and knockout (KO) mice were challenged with a high-fat diet to induce early-stage type 2 diabetes. After 16 wk on the high-fat diet, fasting blood glucose values were increased and glucose tolerance was impaired in the male WT mice. Both of these effects were significantly blunted in the male KO mice. In addition, male and female WT mice developed diabetes-induced hyperfiltration, which was attenuated in the Olfr1393 KO mice and corresponded with a reduction in luminal expression of Sglt2. Collectively, these data indicate that renal Olfr1393 can contribute to the progression of type 2 diabetes, likely as a regulator of Na+-glucose cotransport in the proximal tubule.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Dieta Hiperlipídica , Túbulos Renais Proximais/metabolismo , Obesidade/complicações , Receptores Odorantes/metabolismo , Reabsorção Renal , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Taxa de Filtração Glomerular , Resistência à Insulina , Túbulos Renais Proximais/fisiopatologia , Masculino , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/fisiologia , Fatores de Tempo
13.
Am J Physiol Renal Physiol ; 317(1): F172-F186, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042061

RESUMO

The kidney uses specialized G protein-coupled receptors, including olfactory receptors (ORs), to act as sensors of molecules and metabolites. In the present study, we cloned and studied seven renal ORs, which we previously found to be expressed in the murine renal cortex. As most ORs are orphan receptors, our goal was to identify ligands for these ORs in the hope that this will guide future research into their functional roles. We identified novel ligands for two ORs: Olfr558 and Olfr90. For Olfr558, we confirmed activation by previously reported ligands and identified 16 additional carboxylic acids that activated this OR. The strongest activation of Olfr558 was produced by butyric, cyclobutanecarboxylic, isovaleric, 2-methylvaleric, 3-methylvaleric, 4-methylvaleric, and valeric acids. The primary in vivo source of both butyric and isovaleric acids is gut microbial metabolism. We also identified 14 novel ligands that activated Olfr90, the strongest of which were 2-methyl-4-propyl-1,3-oxathiane, 1-octen-3-ol, 2-octanol, and 3-octanol. Interestingly, 8 of these 14 ligands are of fungal origin. We also investigated the tissue distribution of these receptors and found that they are each found in a subset of "nonsensory" tissues. Finally, we examined the putative human orthologs of Olfr558 and Olfr90 and found that the human ortholog of Olfr558 (OR51E1) has a similar ligand profile, indicating that the role of this OR is likely evolutionarily conserved. In summary, we examined seven novel renal ORs and identified new ligands for Olfr558 and Olfr90, which imply that both of these receptors serve to detect metabolites produced by microorganisms.


Assuntos
Córtex Renal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Odorantes/metabolismo , Animais , Ácidos Carboxílicos/metabolismo , Ácidos Carboxílicos/farmacologia , Microbioma Gastrointestinal , Humanos , Córtex Renal/efeitos dos fármacos , Ligantes , Camundongos Endogâmicos C57BL , Transporte Proteico , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Odorantes/agonistas , Receptores Odorantes/genética , Transdução de Sinais , Distribuição Tecidual
14.
FASEB J ; 32(4): 2046-2059, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29196502

RESUMO

The investigation of orphan GPCRs (GPRs) has the potential to uncover novel insights into whole animal physiology. In this study, our goal was to determine the renal localization of Gprc5c, a receptor that we previously reported to be highly expressed in murine whole kidney, and to examine physiologic parameters in Gprc5c knockout (KO) mice to gain insight into function. Gprc5c localized to the apical membrane of renal proximal tubules (PTs) in mice, rats, and humans. With the comparison of Gprc5c wild-type (WT) and KO mice, we found that Gprc5c KO mice have altered acid-base homeostasis. Specifically, Gprc5c KO mice have lower blood pH and higher urine pH compared with WT mice, with a reduced level of titratable acids in their urine. In an in vitro GPCR internalization assay, we observed that Gprc5c internalization (an index of activation) was triggered by alkaline extracellular pH. Furthermore, with the use of an in vitro BCECF assay, we observed that Gprc5c increases Na+/H+ exchanger 3 (NHE3) activity at alkaline pH. We also find that the NHE3 activity is reduced in Gprc5c KO mice by 2 photon imaging in seminaphthorhodafluors (SNARF)-4F-loaded kidney sections. NHE3 is a primary contributor to apical transport of H+ in the renal PT. Together, these data imply that Gprc5c modulates the renal contribution to systemic pH homeostasis, at least in part, by taking part in the regulation of NHE3.-Rajkumar, P., Cha, B., Yin, J., Arend, L. J., Paunescu, T. G., Hirabayashi, Y., Donowitz, M., Pluznick, J. L. Identifying the localization and exploring a functional role for Gprc5c in the kidney.


Assuntos
Túbulos Renais Proximais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ácidos/sangue , Ácidos/urina , Álcalis/sangue , Álcalis/urina , Animais , Células HEK293 , Humanos , Túbulos Renais Proximais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Receptores Acoplados a Proteínas G/genética , Trocador 3 de Sódio-Hidrogênio/metabolismo , Equilíbrio Hidroeletrolítico
15.
Am J Physiol Renal Physiol ; 315(5): F1187-F1190, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30066586

RESUMO

The kidneys play a critical role in precisely regulating the composition of the plasma to maintain homeostasis. To achieve this, the kidneys must be able to accurately determine or "sense" the concentration of a wide variety of substances and to make adjustments accordingly. Kidneys face a key challenge in the arena of pH balance, as there is a particularly narrow range over which plasma pH varies in a healthy subject (7.35-7.45) and this pH must constantly be protected against a variety of onslaughts (changes in diet, activity, and even elevation). The proximal tubule, the first segment to come into contact with the forming urine, plays an important role in helping the kidneys to maintain pH homeostasis. Recent studies have identified a number of novel proximal tubule proteins and signaling pathways that work to sense changes in pH and subsequently modulate renal pH regulation. In this review, we will highlight the role of novel players in acid-base homeostasis in the proximal tubule.


Assuntos
Equilíbrio Ácido-Base , Túbulos Renais Proximais/metabolismo , Eliminação Renal , Reabsorção Renal , Animais , Sangue/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Receptores de Detecção de Cálcio/metabolismo
16.
Am J Physiol Renal Physiol ; 314(4): F517-F530, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141937

RESUMO

Obesity is a significant risk factor for both chronic kidney disease and end-stage renal disease. To better understand disease development, we sought to identify novel genes differentially expressed early in disease progression. We first confirmed that mice fed a high-fat (HF) diet exhibit early signs of renal injury including hyperfiltration. We then performed RNA-Seq using renal cortex RNA from C57BL6/J male mice fed either HF or control (Ctrl) diet. We identified 1,134 genes differentially expressed in the cortex on HF vs. Ctrl, of which 31 genes were selected for follow-up analysis. This included the 9 most upregulated, the 11 most downregulated, and 11 genes of interest (primarily sensory receptors and G proteins). Quantitative (q)RT-PCR for these 31 genes was performed on additional male renal cortex and medulla samples, and 11 genes (including all 9 upregulated genes) were selected for further study based on qRT-PCR. We then examined expression of these 11 genes in Ctrl and HF male heart and liver samples, which demonstrated that these changes are relatively specific to the renal cortex. These 11 genes were also examined in female renal cortex, where we found that the expression changes seen in males on a HF diet are not replicated in females, even when the females are started on the diet sooner to match weight gain of the males. In sum, these data demonstrate that in a HF-diet model of early disease, novel transcriptional changes occur that are both sex specific and specific to the renal cortex.


Assuntos
Dieta Hiperlipídica , Córtex Renal/metabolismo , Nefropatias/genética , Obesidade/genética , Transcriptoma , Animais , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Córtex Renal/patologia , Nefropatias/etiologia , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Reação em Cadeia da Polimerase , Fatores Sexuais , Fatores de Tempo
18.
Am J Physiol Renal Physiol ; 313(1): F55-F61, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28356283

RESUMO

Glucose homeostasis is highly controlled, and the function of the kidney plays an integral role in this process. The exquisite control of blood glucose relies, in part, on renal glucose filtration, renal glucose reabsorption, and renal gluconeogenesis. Particularly critical to maintaining glucose homeostasis is the renal reabsorption of glucose; with ~162 g of glucose filtered by the kidney per day, it is imperative that the kidney have the ability to efficiently reabsorb nearly 100% of this glucose back in the bloodstream. In this review, we focus on this central process, highlighting the renal transporters and regulators involved in both the physiology and pathophysiology of glucose reabsorption.


Assuntos
Glicemia/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Reabsorção Renal , Animais , Biomarcadores/sangue , Gluconeogênese , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Homeostase , Humanos , Rim/fisiopatologia , Nefropatias/sangue , Nefropatias/fisiopatologia , Proteínas de Transporte de Sódio-Glucose/metabolismo
19.
Curr Hypertens Rep ; 19(4): 25, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28315048

RESUMO

PURPOSE OF REVIEW: Microbial short-chain fatty acids (SCFAs) are byproducts of microbial metabolism which can be absorbed into the bloodstream of the host, where they exert effects on host physiology. SCFAs have been known to influence several aspects of host physiology, including the regulation of blood pressure. In this review, we will consider recent studies linking SCFAs to blood pressure regulation. RECENT FINDINGS: Several recent studies have found that changes in blood pressure often coordinate with expected changes in SCFAS. Efforts are now well underway to dissect and better understand this potential connection. One way that SCFAs can influence host cells is by interacting with host GPCRs, including Gpr41 and Olfr78, among others. Intriguingly, mice null for Olfr78 are hypotensive, whereas mice null for Gpr41 are hypertensive, implying that these pathways may be physiologically important links between SCFAs and host blood pressure control. In sum, these studies demonstrate that there does indeed appear to be a link between SCFAs and blood pressure, which likely involves host GPCRs, at least in part; however, the details and intricacies of these interactions are not yet fully understood and will greatly benefit from further studies.


Assuntos
Pressão Sanguínea , Ácidos Graxos Voláteis/metabolismo , Animais , Humanos , Hipertensão
20.
Physiol Genomics ; 48(11): 826-834, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27664183

RESUMO

Short chain fatty acid (SCFA) metabolites are byproducts of gut microbial metabolism that are known to affect host physiology via host G protein-coupled receptor (GPCRs). We previously showed that an acute SCFA bolus decreases blood pressure (BP) in anesthetized mice, an effect mediated primarily via Gpr41. In this study, our aims were to identify the cellular localization of Gpr41 and to determine its role in BP regulation. We localized Gpr41 to the vascular endothelium using RT-PCR: Gpr41 is detected in intact vessels (with endothelium) but is absent from denuded vessels (without endothelium). Furthermore, using pressure myography we confirmed that SCFAs dilate resistance vessels in an endothelium-dependent manner. Since we previously found that Gpr41 mediates a hypotensive response to acute SCFA administration, we hypothesized that Gpr41 knockout (KO) mice would be hypertensive. Here, we report that Gpr41 KO mice have isolated systolic hypertension compared with wild-type (WT) mice; diastolic BP was not different between WT and KO. Older Gpr41 KO mice also exhibited elevated pulse wave velocity, consistent with a phenotype of systolic hypertension; however, there was no increase in ex vivo aorta stiffness (measured by mechanical tensile testing). Plasma renin concentrations were also similar in KO and WT mice. The systolic hypertension in Gpr41 KO is not salt sensitive, as it is not significantly altered on either a high- or low-salt diet. In sum, these studies suggest that endothelial Gpr41 lowers baseline BP, likely by decreasing active vascular tone without altering passive characteristics of the blood vessels, and that Gpr41 KO mice have hypertension of a vascular origin.


Assuntos
Bactérias/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Endotélio Vascular/metabolismo , Ácidos Graxos Voláteis/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Feminino , Hipertensão/sangue , Hipertensão/fisiopatologia , Masculino , Metaboloma/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Renina/sangue , Cloreto de Sódio na Dieta/efeitos adversos , Sus scrofa , Sístole/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa