Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Physiol ; 602(4): 737-757, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38345534

RESUMO

Paraoxonase 3 (PON3) is expressed in the aldosterone-sensitive distal nephron, where filtered Na+ is reabsorbed mainly via the epithelial Na+ channel (ENaC) and Na+ -coupled co-transporters. We previously showed that PON3 negatively regulates ENaC through a chaperone mechanism. The present study aimed to determine the physiological role of PON3 in renal Na+ and K+ homeostasis. Pon3 knockout (KO) mice had higher amiloride-induced natriuresis and lower plasma [K+ ] at baseline. Single channel recordings in split-open tubules showed that the number of active channels per patch was significantly higher in KO mice, resulting in a higher channel activity in the absence of PON3. Although whole kidney abundance of ENaC subunits was not altered in Pon3 KOs, ENaC gamma subunit was more apically distributed within the connecting tubules and cortical collecting ducts of Pon3 KO kidneys. Additionally, small interfering RNA-mediated knockdown of PON3 in cultured mouse cortical collecting duct cells led to an increased surface abundance of ENaC gamma subunit. As a result of lower plasma [K+ ], sodium chloride co-transporter phosphorylation was enhanced in the KO kidneys, a phenotype that was corrected by a high K+ diet. Finally, PON3 expression was upregulated in mouse kidneys under dietary K+ restriction, potentially providing a mechanism to dampen ENaC activity and associated K+ secretion. Taken together, our results show that PON3 has a role in renal Na+ and K+ homeostasis through regulating ENaC functional expression in the distal nephron. KEY POINTS: Paraoxonase 3 (PON3) is expressed in the distal nephron of mouse kidneys and functions as a molecular chaperone to reduce epithelial Na+ channel (ENaC) expression and activity in heterologous expression systems. We examined the physiological role of PON3 in renal Na+ and K+ handling using a Pon3 knockout (KO) mouse model. At baseline, Pon3 KO mice had lower blood [K+ ], more functional ENaC in connecting tubules/cortical collecting ducts, higher amiloride-induced natriuresis, and enhanced sodium chloride co-transporter (NCC) phosphorylation. Upon challenge with a high K+ diet, Pon3 KO mice had normalized blood [K+ ] and -NCC phosphorylation but lower circulating aldosterone levels compared to their littermate controls. Kidney PON3 abundance was altered in mice under dietary K+ loading or K+ restriction, providing a potential mechanism for regulating ENaC functional expression and renal Na+ and K+ homeostasis in the distal nephron.


Assuntos
Amilorida , Simportadores , Camundongos , Animais , Amilorida/farmacologia , Arildialquilfosfatase/metabolismo , Canais Epiteliais de Sódio/metabolismo , Aldosterona/metabolismo , Cloreto de Sódio/metabolismo , Sódio/metabolismo , Néfrons/metabolismo
2.
J Physiol ; 602(5): 967-987, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38294810

RESUMO

Aldosterone is responsible for maintaining volume and potassium homeostasis. Although high salt consumption should suppress aldosterone production, individuals with hyperaldosteronism lose this regulation, leading to a state of high aldosterone despite dietary sodium consumption. The present study examines the effects of elevated aldosterone, with or without high salt consumption, on the expression of key Na+ transporters and remodelling in the distal nephron. Epithelial sodium channel (ENaC) α-subunit expression was increased with aldosterone regardless of Na+ intake. However, ENaC ß- and γ-subunits unexpectedly increased at both a transcript and protein level with aldosterone when high salt was present. Expression of total and phosphorylated Na+ Cl- cotransporter (NCC) significantly increased with aldosterone, in association with decreased blood [K+ ], but the addition of high salt markedly attenuated the aldosterone-dependent NCC increase, despite equally severe hypokalaemia. We hypothesized this was a result of differences in distal convoluted tubule length when salt was given with aldosterone. Imaging and measurement of the entire pNCC-positive tubule revealed that aldosterone alone caused a shortening of this segment, although the tubule had a larger cross-sectional diameter. This was not true when salt was given with aldosterone because the combination was associated with a lengthening of the tubule in addition to increased diameter, suggesting that differences in the pNCC-positive area are not responsible for differences in NCC expression. Together, our results suggest the actions of aldosterone, and the subsequent changes related to hypokalaemia, are altered in the presence of high dietary Na+ . KEY POINTS: Aldosterone regulates volume and potassium homeostasis through effects on transporters in the kidney; its production can be dysregulated, preventing its suppression by high dietary sodium intake. Here, we examined how chronic high sodium consumption affects aldosterone's regulation of sodium transporters in the distal nephron. Our results suggest that high sodium consumption with aldosterone is associated with increased expression of all three epithelial sodium channel subunits, rather than just the alpha subunit. Aldosterone and its associated decrease in blood [K+ ] lead to an increased expression of Na-Cl cotransporter (NCC); the addition of high sodium consumption with aldosterone partially attenuates this NCC expression, despite similarly low blood [K+ ]. Upstream kinase regulators and tubule remodelling do not explain these results.


Assuntos
Hipopotassemia , Sódio na Dieta , Humanos , Sódio na Dieta/farmacologia , Sódio na Dieta/metabolismo , Sódio/metabolismo , Aldosterona/farmacologia , Aldosterona/metabolismo , Canais Epiteliais de Sódio/metabolismo , Hipopotassemia/metabolismo , Túbulos Renais Distais/metabolismo , Cloreto de Sódio na Dieta , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Potássio/metabolismo
3.
Am J Physiol Renal Physiol ; 327(1): F49-F60, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38779757

RESUMO

The pore-forming α-subunit of the large-conductance K+ (BK) channel is encoded by a single gene, KCNMA1. BK channel-mediated K+ secretion in the kidney is crucial for overall renal K+ homeostasis in both physiological and pathological conditions. BK channels achieve phenotypic diversity by various mechanisms, including substantial exon rearrangements at seven major alternative splicing sites. However, KCNMA1 alternative splicing in the kidney has not been characterized. The present study aims to identify the major splice variants of mouse Kcnma1 in whole kidney and distal nephron segments. We designed primers that specifically cross exons within each alternative splice site of mouse Kcnma1 and performed real-time quantitative RT-PCR (RT-qPCR) to quantify relative abundance of each splice variant. Our data suggest that Kcnma1 splice variants within mouse kidney are less diverse than in the brain. During postnatal kidney development, most Kcnma1 splice variants at site 5 and the COOH terminus increase in abundance over time. Within the kidney, the regulation of Kcnma1 alternative exon splicing within these two sites by dietary K+ loading is both site and sex specific. In microdissected distal tubules, the Kcnma1 alternative splicing profile, as well as its regulation by dietary K+, are distinctly different than in the whole kidney, suggesting segment and/or cell type specificity in Kcnma1 splicing events. Overall, our data provide evidence that Kcnma1 alternative splicing is regulated during postnatal development and may serve as an important adaptive mechanism to dietary K+ loading in mouse kidney.NEW & NOTEWORTHY We identified the major Kcnma1 splice variants that are specifically expressed in the whole mouse kidney or aldosterone-sensitive distal nephron segments. Our data suggest that Kcnma1 alternative splicing is developmentally regulated and subject to changes in dietary K+.


Assuntos
Processamento Alternativo , Rim , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Potássio na Dieta , Animais , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Potássio na Dieta/metabolismo , Rim/metabolismo , Camundongos Endogâmicos C57BL , Camundongos , Masculino , Regulação da Expressão Gênica no Desenvolvimento , Éxons , Feminino
4.
Am J Physiol Renal Physiol ; 321(2): F135-F148, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34151589

RESUMO

Cell-associated kidney injury molecule-1 (KIM-1) exerts an anti-inflammatory role following kidney injury by mediating efferocytosis and downregulating the NF-κB pathway. KIM-1 cleavage blunts its anti-inflammatory activities. We reported that mucin 1 (MUC1) is protective in a mouse model of ischemia-reperfusion injury (IRI). As both KIM-1 and MUC1 are induced in the proximal tubule (PT) during IRI and are a disintegrin and metalloprotease 17 (ADAM17) substrates, we tested the hypothesis that MUC1 protects KIM-1 activity. Muc1 knockout (KO) mice and wild-type (WT) littermates were subjected to IRI. KIM-1, MUC1, and ADAM17 levels (and signaling pathways) were assessed by immunoblot analysis. PT localization was assessed by confocal microscopy and an in situ proximity ligation assay. Findings were extended using human kidneys and urine as well as KIM-1-mediated efferocytosis assays in mouse PT cultures. In response to tubular injury in mouse and human kidneys, we observed induction and coexpression of KIM-1 and MUC1 in the PT. Compared with WT mice, Muc1 KO mice had higher urinary KIM-1 and lower kidney KIM-1. KIM-1 was apical in the PT of WT kidneys but predominately with luminal debris in Muc1 KO mice. Efferocytosis was reduced in Muc1 KO PT cultures compared with WT cultures, whereas inflammation was increased in Muc1 KO kidneys compared with WT kidneys. MUC1 was cleaved by ADAM17 in PT cultures and blocked KIM-1 shedding in Madin-Darby canine kidney cells. We conclude that KIM-1-mediated efferocytosis and thus anti-inflammatory activity during IRI is preserved in the injured kidney by MUC1 inhibition of KIM-1 shedding.NEW & NOTEWORTHY KIM-1 plays a key role in the recovery of the tubule epithelium during renal IRI by mediating efferocytosis and associated signaling that suppresses inflammation. Excessive cleavage of KIM-1 by ADAM17 provides a decoy receptor that aggravates efferocytosis and subsequent signaling. Our data from experiments in mice, patients, and cultured cells show that MUC1 is also induced during IRI and competes with KIM-1 for cleavage by ADAM17. Consequently, MUC1 protects KIM-1 anti-inflammatory activity in the damaged kidney.


Assuntos
Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Inflamação/metabolismo , Túbulos Renais Proximais/metabolismo , Rim/irrigação sanguínea , Mucina-1/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteína ADAM17/metabolismo , Animais , Linhagem Celular , Cães , Humanos , Rim/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Mucina-1/genética , Fagocitose/fisiologia
5.
Int J Mol Sci ; 22(20)2021 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-34681576

RESUMO

Acute kidney injury due to renal ischemia-reperfusion injury (IRI) may lead to chronic or end stage kidney disease. A greater understanding of the cellular mechanisms underlying IRI are required to develop therapeutic options aimed at limiting or reversing damage from IRI. Prior work has shown that deletion of the α subunit of the epithelial Na+ channel (ENaC) in endothelial cells protects from IRI by increasing the availability of nitric oxide. While canonical ENaCs consist of an α, ß, and γ subunit, there is evidence of non-canonical ENaC expression in endothelial cells involving the α subunit. We therefore tested whether the deletion of the γ subunit of ENaC also protects mice from IRI to differentiate between these channel configurations. Mice with endothelial-specific deletion of the γ subunit and control littermates were subjected to unilateral renal artery occlusion followed by 48 h of reperfusion. No significant difference was noted in injury between the two groups as assessed by serum creatinine and blood urea nitrogen, levels of specific kidney injury markers, and histological examination. While deletion of the γ subunit did not alter infiltration of immune cells or cytokine message, it was associated with an increase in levels of total and phosphorylated endothelial nitric oxide synthase (eNOS) in the injured kidneys. Our studies demonstrate that even though deletion of the γ subunit of ENaC may allow for greater activation of eNOS, this is not sufficient to prevent IRI, suggesting the protective effects of α subunit deletion may be due, in part, to other mechanisms.


Assuntos
Injúria Renal Aguda/genética , Células Endoteliais/metabolismo , Canais Epiteliais de Sódio/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Traumatismo por Reperfusão/genética , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Animais , Nitrogênio da Ureia Sanguínea , Estudos de Casos e Controles , Linhagem Celular , Creatinina/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Canais Epiteliais de Sódio/metabolismo , Deleção de Genes , Masculino , Camundongos , Fosforilação , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo
6.
Am J Physiol Renal Physiol ; 318(6): F1341-F1356, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32281415

RESUMO

We characterized mouse blood pressure and ion transport in the setting of commonly used rodent diets that drive K+ intake to the extremes of deficiency and excess. Male 129S2/Sv mice were fed either K+-deficient, control, high-K+ basic, or high-KCl diets for 10 days. Mice maintained on a K+-deficient diet exhibited no change in blood pressure, whereas K+-loaded mice developed an ~10-mmHg blood pressure increase. Following challenge with NaCl, K+-deficient mice developed a salt-sensitive 8 mmHg increase in blood pressure, whereas blood pressure was unchanged in mice fed high-K+ diets. Notably, 10 days of K+ depletion induced diabetes insipidus and upregulation of phosphorylated NaCl cotransporter, proximal Na+ transporters, and pendrin, likely contributing to the K+-deficient NaCl sensitivity. While the anionic content with high-K+ diets had distinct effects on transporter expression along the nephron, both K+ basic and KCl diets had a similar increase in blood pressure. The blood pressure elevation on high-K+ diets correlated with increased Na+-K+-2Cl- cotransporter and γ-epithelial Na+ channel expression and increased urinary response to furosemide and amiloride. We conclude that the dietary K+ maneuvers used here did not recapitulate the inverse effects of K+ on blood pressure observed in human epidemiological studies. This may be due to the extreme degree of K+ stress, the low-Na+-to-K+ ratio, the duration of treatment, and the development of other coinciding events, such as diabetes insipidus. These factors must be taken into consideration when studying the physiological effects of dietary K+ loading and depletion.


Assuntos
Pressão Arterial , Hipertensão/metabolismo , Túbulos Renais/metabolismo , Deficiência de Potássio/metabolismo , Potássio na Dieta/metabolismo , Cloreto de Sódio na Dieta/metabolismo , Ração Animal , Animais , Diabetes Insípido/etiologia , Diabetes Insípido/metabolismo , Diabetes Insípido/fisiopatologia , Canais Epiteliais de Sódio/metabolismo , Hipertensão/etiologia , Hipertensão/fisiopatologia , Transporte de Íons , Túbulos Renais/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Natriurese , Fosforilação , Deficiência de Potássio/etiologia , Deficiência de Potássio/fisiopatologia , Potássio na Dieta/administração & dosagem , Potássio na Dieta/toxicidade , Simportadores de Cloreto de Sódio/metabolismo , Cloreto de Sódio na Dieta/toxicidade , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Transportadores de Sulfato/metabolismo
7.
J Biol Chem ; 293(27): 10757-10766, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29743244

RESUMO

The Caenorhabditis elegans MEC-4/MEC-10 channel mediates the worm's response to gentle body touch and is activated by laminar shear stress (LSS) when expressed in Xenopus oocytes. Substitutions at multiple sites within the second transmembrane domain (TM2) of MEC-4 or MEC-10 abolish the gentle touch response in worms, but the roles of these residues in mechanosensing are unclear. The present study therefore examined the role of specific MEC-4 and MEC-10 TM2 residues in the channel's response to LSS. We found that introducing mutations within the TM2 of MEC-4 or MEC-10 not only altered channel activity, but also affected the channel's response to LSS. This response was enhanced by Cys substitutions at selected MEC-4 sites (Phe715, Gly716, Gln718, and Leu719) between the degenerin and the putative amiloride-binding sites in this subunit. In contrast, the LSS response was largely blunted in MEC-10 variants bearing single Cys substitutions in the regions preceding and following the amiloride-binding site (Gly677-Leu681), as well as with four MEC-10 touch-deficient mutations that introduced charged residues into the TM2 domain. An enhanced response to LSS was observed with a MEC-10 mutation in the putative selectivity filter. Overall, MEC-4 or MEC-10 mutants that altered the channel's LSS response are primarily clustered between the degenerin site and the selectivity filter, a region that probably forms the narrowest portion of the channel pore. Our results suggest that pore-lining residues of MEC-4 and MEC-10 have important yet different roles in tuning the channel's response to mechanical forces.


Assuntos
Aminoácidos/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Membrana/metabolismo , Estresse Mecânico , Xenopus laevis/metabolismo , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Células Cultivadas , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Mutação , Oócitos/citologia , Oócitos/metabolismo , Conformação Proteica , Homologia de Sequência , Xenopus laevis/crescimento & desenvolvimento
8.
Curr Opin Nephrol Hypertens ; 28(2): 113-119, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30585851

RESUMO

PURPOSE OF REVIEW: This review describes recent findings regarding the epithelial Na channel (ENaC) and its roles in physiologic and pathophysiologic states. We discuss new insights regarding ENaC's structure, its regulation by various factors, its potential role in hypertension and nephrotic syndrome, and its roles in the immune system and vasculature. RECENT FINDINGS: A recently resolved structure of ENaC provides clues regarding mechanisms of ENaC activation by proteases. The use of amiloride in nephrotic syndrome, and associated complications are discussed. ENaC is expressed in dendritic cells and contributes to immune system activation and increases in blood pressure in response to NaCl. ENaC is expressed in endothelial ENaC and has a role in regulating vascular tone. SUMMARY: New findings have emerged regarding ENaC and its role in the kidney, immune system, and vasculature.


Assuntos
Endotélio/metabolismo , Canais Epiteliais de Sódio/metabolismo , Hipertensão/metabolismo , Síndrome Nefrótica/metabolismo , Animais , Pressão Sanguínea , Vasos Sanguíneos/fisiologia , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/fisiologia , Humanos , Sistema Imunitário/metabolismo
9.
Microcirculation ; 24(8)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28857417

RESUMO

OBJECTIVE: Previously, we found that diet-induced HHcy in mice caused decreased eNOS expression and signaling in mesenteric arteries, but greatly enhanced non-NOS, non-prostacyclin-dependent vasodilation, which involves MEJ communication. To further assess whether HHcy enhances MEJ communication, this study examined endothelium-dependent attenuation of phenylephrine-induced vasoconstriction (myoendothelial feedback) and key molecules involved. METHODS: Myoendothelial feedback was examined in isolated mouse mesenteric arteries, after 6-weeks diet-induced HHcy, using pressure myography. Gap junction (Cx37, Cx40, Cx43), NOS (eNOS, nNOS, iNOS), and potassium channel (IK1) protein expression were measured with immunoblots, and connexin mRNAs with real-time PCR. Contribution of nNOS + iNOS to vasomotor responses was assessed using the drug TRIM. RESULTS: Myoendothelial feedback was significantly (P < .05) enhanced in HHcy arteries compared to control, coincident with significantly greater Cx37 and IK1 protein and Cx37 mRNA. Cx43 protein, but not mRNA, was significantly less in HHcy, and Cx40 was not different. eNOS protein was significantly less in HHcy. nNOS and iNOS were not different. TRIM had little effect on vasomotor function. CONCLUSIONS: Diet-induced HHcy enhanced myoendothelial feedback, and increased Cx37 and IK1 expression may contribute. nNOS or iNOS did not upregulate to compensate for decreased eNOS, and they had little involvement in vasomotor function.


Assuntos
Conexinas/metabolismo , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica , Hiper-Homocisteinemia/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/biossíntese , Artérias Mesentéricas/metabolismo , Animais , Alimentos Formulados/efeitos adversos , Junções Comunicantes/patologia , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia , Masculino , Artérias Mesentéricas/patologia , Artérias Mesentéricas/fisiopatologia , Camundongos , Óxido Nítrico Sintase Tipo I/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo III/biossíntese , Proteína alfa-4 de Junções Comunicantes
10.
Vasc Med ; 22(5): 363-369, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28825355

RESUMO

Hemoglobin subunit alpha (HBA) expression in endothelial cells (ECs) has recently been shown to control vascular tone and function. We sought to elucidate the transcriptional regulation of HBA expression in the EC. Gain of KLF2 or KLF4 function studies led to significant induction of HBA in ECs. An opposite effect was observed in ECs isolated from animals with endothelial-specific ablation of Klf2, Klf4 or both. Promoter reporter assays demonstrated that KLF2/KLF4 transactivated the hemoglobin alpha promoter, an effect that was abrogated following mutation of all four putative KLF-binding sites. Fine promoter mutational studies localized three out of four KLF-binding sites (sites 2, 3, and 4) as critical for the transactivation of the HBA promoter by KLF2/KLF4. Chromatin immunoprecipitation studies showed that KLF4 bound to the HBA promoter in ECs. Thus, KLF2 and KLF4 serve as important regulators that promote HBA expression in the endothelium.


Assuntos
Células Endoteliais/metabolismo , Hemoglobinas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Sítios de Ligação , Bovinos , Células Cultivadas , Genótipo , Hemoglobinas/genética , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fragmentos de Peptídeos/genética , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica , Ativação Transcricional , Transfecção , Regulação para Cima
11.
J Biol Chem ; 290(27): 16861-72, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-26001785

RESUMO

NADH cytochrome b5 reductase 3 (CYB5R3) is critical for reductive reactions such as fatty acid elongation, cholesterol biosynthesis, drug metabolism, and methemoglobin reduction. Although the physiological and metabolic importance of CYB5R3 has been established in hepatocytes and erythrocytes, emerging investigations suggest that CYB5R3 is critical for nitric oxide signaling and vascular function. However, advancement toward fully understanding CYB5R3 function has been limited due to a lack of potent small molecule inhibitors. Because of this restriction, we modeled the binding mode of propylthiouracil, a weak inhibitor of CYB5R3 (IC50 = ∼275 µM), and used it as a guide to predict thiouracil-biased inhibitors from the set of commercially available compounds in the ZINC database. Using this approach, we validated two new potent derivatives of propylthiouracil, ZINC05626394 (IC50 = 10.81 µM) and ZINC39395747 (IC50 = 9.14 µM), both of which inhibit CYB5R3 activity in cultured cells. Moreover, we found that ZINC39395747 significantly increased NO bioavailability in renal vascular cells, augmented renal blood flow, and decreased systemic blood pressure in response to vasoconstrictors in spontaneously hypertensive rats. These compounds will serve as a new tool to examine the biological functions of CYB5R3 in physiology and disease and also as a platform for new drug development.


Assuntos
Citocromo-B(5) Redutase/química , Inibidores Enzimáticos/química , Óxido Nítrico/metabolismo , Propiltiouracila/química , Animais , Citocromo-B(5) Redutase/genética , Citocromo-B(5) Redutase/metabolismo , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Humanos , Estrutura Molecular , Propiltiouracila/metabolismo , Ratos
12.
Nitric Oxide ; 49: 8-15, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26028569

RESUMO

Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.


Assuntos
Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Resistência Vascular/fisiologia , Animais , Conexinas/metabolismo , Retroalimentação Fisiológica/fisiologia , Hemoglobinas/metabolismo , Humanos , Camundongos
13.
Arterioscler Thromb Vasc Biol ; 34(12): 2594-600, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25278292

RESUMO

OBJECTIVE: Hemoglobin α (Hb α) and endothelial nitric oxide synthase (eNOS) form a macromolecular complex at myoendothelial junctions; the functional role of this interaction remains undefined. To test if coupling of eNOS and Hb α regulates nitric oxide signaling, vascular reactivity, and blood pressure using a mimetic peptide of Hb α to disrupt this interaction. APPROACH AND RESULTS: In silico modeling of Hb α and eNOS identified a conserved sequence of interaction. By mutating portions of Hb α, we identified a specific sequence that binds eNOS. A mimetic peptide of the Hb α sequence (Hb α X) was generated to disrupt this complex. Using in vitro binding assays with purified Hb α and eNOS and ex vivo proximity ligation assays on resistance arteries, we have demonstrated that Hb α X significantly decreased interaction between eNOS and Hb α. Fluorescein isothiocyanate labeling of Hb α X revealed localization to holes in the internal elastic lamina (ie, myoendothelial junctions). To test the functional effects of Hb α X, we measured cyclic guanosine monophosphate and vascular reactivity. Our results reveal augmented cyclic guanosine monophosphate production and altered vasoconstriction with Hb α X. To test the in vivo effects of these peptides on blood pressure, normotensive and hypertensive mice were injected with Hb α X, which caused a significant decrease in blood pressure; injection of Hb α X into eNOS(-/-) mice had no effect. CONCLUSIONS: These results identify a novel sequence on Hb α that is important for Hb α/eNOS complex formation and is critical for nitric oxide signaling at myoendothelial junctions.


Assuntos
Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Vasoconstrição/fisiologia , alfa-Globinas/metabolismo , Sequência de Aminoácidos , Animais , Pressão Sanguínea/fisiologia , Células Cultivadas , Simulação por Computador , Sequência Conservada , Células Endoteliais/metabolismo , Humanos , Junções Intercelulares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Resistência Vascular/fisiologia , alfa-Globinas/química , alfa-Globinas/genética
14.
J Clin Invest ; 134(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38426496

RESUMO

Ca2+-activated BK channels in renal intercalated cells (ICs) mediate luminal flow-induced K+ secretion (FIKS), but how ICs sense increased flow remains uncertain. We examined whether PIEZO1, a mechanosensitive Ca2+-permeable channel expressed in the basolateral membranes of ICs, is required for FIKS. In isolated cortical collecting ducts (CCDs), the mechanosensitive cation-selective channel inhibitor GsMTx4 dampened flow-induced increases in intracellular Ca2+ concentration ([Ca2+]i), whereas the PIEZO1 activator Yoda1 increased [Ca2+]i and BK channel activity. CCDs from mice fed a high-K+ (HK) diet exhibited a greater Yoda1-dependent increase in [Ca2+]i than CCDs from mice fed a control K+ diet. ICs in CCDs isolated from mice with a targeted gene deletion of Piezo1 in ICs (IC-Piezo1-KO) exhibited a blunted [Ca2+]i response to Yoda1 or increased flow, with an associated loss of FIKS in CCDs. Male IC-Piezo1-KO mice selectively exhibited an increased blood [K+] in response to an oral K+ bolus and blunted urinary K+ excretion following a volume challenge. Whole-cell expression of BKα subunit was reduced in ICs of IC-Piezo1-KO mice fed an HK diet. We conclude that PIEZO1 mediates flow-induced basolateral Ca2+ entry into ICs, is upregulated in the CCD in response to an HK diet, and is necessary for FIKS.


Assuntos
Túbulos Renais Coletores , Masculino , Camundongos , Animais , Túbulos Renais Coletores/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Cálcio/metabolismo , Néfrons/metabolismo , Rim/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo
15.
bioRxiv ; 2024 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-38260467

RESUMO

The maintenance of fluid and electrolyte homeostasis by the kidney requires proper folding and trafficking of ion channels and transporters in kidney epithelia. Each of these processes requires a specific subset of a diverse class of proteins termed molecular chaperones. One such chaperone is GRP170, which is an Hsp70-like, endoplasmic reticulum (ER)-localized chaperone that plays roles in protein quality control and protein folding in the ER. We previously determined that loss of GRP170 in the mouse nephron leads to hypovolemia, electrolyte imbalance, and rapid weight loss. In addition, GRP170-deficient mice develop an AKI-like phenotype, typified by tubular injury, elevation of clinical kidney injury markers, and induction of the unfolded protein response (UPR). By using an inducible GRP170 knockout cellular model, we confirmed that GRP170 depletion induces the UPR, triggers an apoptotic response, and disrupts protein homeostasis. Based on these data, we hypothesized that UPR induction underlies hyponatremia and volume depletion in rodents, but that these and other phenotypes might be rectified by supplementation with high salt. To test this hypothesis, control and GRP170 tubule-specific knockout mice were provided with a diet containing 8% sodium chloride. We discovered that sodium supplementation improved electrolyte imbalance and reduced clinical kidney injury markers, but was unable to restore weight or tubule integrity. These results are consistent with UPR induction contributing to the kidney injury phenotype in the nephron-specific GR170 knockout model, and that the role of GRP170 in kidney epithelia is essential to both maintain electrolyte balance and cellular protein homeostasis.

16.
Physiol Rep ; 11(1): e15554, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36636010

RESUMO

The epithelial Na+ channel (ENaC) is traditionally composed of three subunits, although non-canonical expression has been found in various tissues including the vasculature, brain, lung, and dendritic cells of the immune system. Studies of ENaC structure and function have largely relied on heterologous expression systems, often with epitope-tagged channel subunits. Relevant in vivo physiological studies have used ENaC inhibitors, mice with global or tissue specific knockout of subunits, and anti-ENaC subunit antibodies generated by investigators or by commercial sources. Availability of well-characterized, specific antibodies is imperative as we move forward in understanding the role of ENaC in non-epithelial tissues where expression, subunit organization, and electrophysiological characteristics may differ from epithelial tissues. We report that a commonly used commercial anti-α subunit antibody recognizes an intense non-specific band on mouse whole kidney and lung immunoblots, which migrates adjacent to a less intense, aldosterone-induced full length α-subunit. This antibody localizes to the basolateral membrane of aquaporin 2 negative cells in kidney medulla. We validated antibodies against the ß- and γ-subunits from the same commercial source. Our work illustrates the importance of validation studies when using popular, commercially available anti-ENaC antibodies.


Assuntos
Canais Epiteliais de Sódio , Rim , Camundongos , Animais , Canais Epiteliais de Sódio/metabolismo , Rim/metabolismo , Sódio/metabolismo , Epitélio/metabolismo , Medula Renal/metabolismo
17.
JCI Insight ; 8(21)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37707951

RESUMO

Epithelial Na+ channels (ENaCs) control extracellular fluid volume by facilitating Na+ absorption across transporting epithelia. In vitro studies showed that Cys-palmitoylation of the γENaC subunit is a major regulator of channel activity. We tested whether γ subunit palmitoylation sites are necessary for channel function in vivo by generating mice lacking the palmitoylated cysteines (γC33A,C41A) using CRISPR/Cas9 technology. ENaCs in dissected kidney tubules from γC33A,C41A mice had reduced open probability compared with wild-type (WT) littermates maintained on either standard or Na+-deficient diets. Male mutant mice also had higher aldosterone levels than WT littermates following Na+ restriction. However, γC33A,C41A mice did not have reduced amiloride-sensitive Na+ currents in the distal colon or benzamil-induced natriuresis compared to WT mice. We identified a second, larger conductance cation channel in the distal nephron with biophysical properties distinct from ENaC. The activity of this channel was higher in Na+-restricted γC33A,C41A versus WT mice and was blocked by benzamil, providing a possible compensatory mechanism for reduced prototypic ENaC function. We conclude that γ subunit palmitoylation sites are required for prototypic ENaC activity in vivo but are not necessary for amiloride/benzamil-sensitive Na+ transport in the distal nephron or colon.


Assuntos
Amilorida , Lipoilação , Camundongos , Masculino , Animais , Amilorida/farmacologia , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Sódio/metabolismo
18.
bioRxiv ; 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37790468

RESUMO

Sodium and fluid retention in liver disease is classically thought to result from reduced effective circulating volume and stimulation of the renin-angiotensin-aldosterone system (RAAS). Aldosterone dives Na+ retention by activating the mineralocorticoid receptor and promoting the maturation and apical surface expression of the epithelial Na+ channel (ENaC), found in the aldosterone-sensitive distal nephron. However, evidence of fluid retention without RAAS activation suggests the involvement of additional mechanisms. Liver disease can greatly increase plasma and urinary bile acid concentrations and have been shown to activate ENaC in vitro. We hypothesize that elevated bile acids in liver disease activate ENaC and drive fluid retention independent of RAAS. We therefore increased circulating bile acids in mice through bile duct ligation (BDL) and measured effects on urine and body composition, while using spironolactone to antagonize the mineralocorticoid receptor. We found BDL lowered blood [K+] and hematocrit, and increased benzamil-sensitive natriuresis compared to sham, consistent with ENaC activation. BDL mice also gained significantly more body water. Blocking ENaC reversed fluid gains in BDL mice but had no effect in shams. In isolated collecting ducts from rabbits, taurocholic acid stimulated net Na+ absorption but had no effect on K+ secretion or flow-dependent ion fluxes. Our results provide experimental evidence for a novel aldosterone-independent mechanism for sodium and fluid retention in liver disease which may provide additional therapeutic options for liver disease patients.

19.
Physiol Rep ; 10(7): e15255, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35384364

RESUMO

The maintenance of endothelial health is required for normal vascular function and blood pressure regulation. The epithelial Na+ channel (ENaC) in endothelial cells has emerged as a new molecular player in the regulation of endothelial nitric oxide production and vascular stiffness. While ENaC expression in the kidney is negatively regulated by high [Na+ ], ENaC expression in isolated endothelial cells has been shown to increase in response to a high extracellular [Na+ ]. In culture, this increased expression leads to cellular stiffening and decreased nitric oxide release. In vivo, the effects of high salt diet on endothelial ENaC expression and activity have varied depending on the animal model utilized. Our aim in the present study was to examine the role of endothelial ENaC in mediating vasorelaxation in the C57Bl/6 mouse strain. We utilized pressure myography to test the responsiveness of thoracodorsal arteries to acetylcholine in mice with increased sodium consumption both in the presence and absence of increased aldosterone. ENaC's contribution was assessed with the use of the specific inhibitor amiloride. We found that while aldosterone had very little effect on ENaC's contribution to acetylcholine sensitivity, a high salt diet led to an amiloride-dependent shift in the acetylcholine response of vessels. However, the direction of this shift was dependent on the length of high salt diet administration. Overall, our studies reveal that ENaC's role in the endothelium may be more complicated than previously thought. The channel does not simply inhibit nitric oxide generation, but instead helps preserve a homeostatic response.


Assuntos
Amilorida , Vasodilatação , Acetilcolina/farmacologia , Amilorida/farmacologia , Animais , Dieta , Células Endoteliais/metabolismo , Canais Epiteliais de Sódio/metabolismo , Camundongos
20.
JCI Insight ; 7(5)2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35104250

RESUMO

Molecular chaperones are responsible for maintaining cellular homeostasis, and one such chaperone, GRP170, is an endoplasmic reticulum (ER) resident that oversees both protein biogenesis and quality control. We previously discovered that GRP170 regulates the degradation and assembly of the epithelial sodium channel (ENaC), which reabsorbs sodium in the distal nephron and thereby regulates salt-water homeostasis and blood pressure. To define the role of GRP170 - and, more generally, molecular chaperones in kidney physiology - we developed an inducible, nephron-specific GRP170-KO mouse. Here, we show that GRP170 deficiency causes a dramatic phenotype: profound hypovolemia, hyperaldosteronemia, and dysregulation of ion homeostasis, all of which are associated with the loss of ENaC. Additionally, the GRP170-KO mouse exhibits hallmarks of acute kidney injury (AKI). We further demonstrate that the unfolded protein response (UPR) is activated in the GRP170-deficient mouse. Notably, the UPR is also activated in AKI when originating from various other etiologies, including ischemia, sepsis, glomerulonephritis, nephrotic syndrome, and transplant rejection. Our work establishes the central role of GRP170 in kidney homeostasis and directly links molecular chaperone function to kidney injury.


Assuntos
Injúria Renal Aguda , Proteínas de Choque Térmico HSP70 , Animais , Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico HSP70/metabolismo , Camundongos , Chaperonas Moleculares/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA