Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
FASEB J ; 34(6): 7941-7957, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32293069

RESUMEN

Acute renal depletion of sorting nexin 1 (SNX1) in mice results in blunted natriuretic response and hypertension due to impaired dopamine D5 receptor (D5 R) activity. We elucidated the molecular mechanisms for these phenotypes in Snx1-/- mice. These mice had increased renal expressions of angiotensin II type 1 receptor (AT1 R), NADPH oxidase (NOX) subunits, D5 R, and NaCl cotransporter. Basal reactive oxygen species (ROS), NOX activity, and blood pressure (BP) were also higher in Snx1-/- mice, which were normalized by apocynin, a drug that prevents NOX assembly. Renal proximal tubule (RPT) cells from hypertensive (HT) Euro-American males had deficient SNX1 activity, impaired D5 R endocytosis, and increased ROS compared with cells from normotensive (NT) Euro-American males. siRNA-mediated depletion of SNX1 in RPT cells from NT subjects led to a blunting of D5 R agonist-induced increase in cAMP production and decrease in Na+ transport, effects that were normalized by over-expression of SNX1. Among HT African-Americans, three of the 12 single nucleotide polymorphisms interrogated for the SNX1 gene were associated with a decrease in systolic BP in response to hydrochlorothiazide (HCTZ). The results illustrate a new paradigm for the development of hypertension and imply that the trafficking protein SNX1 may be a crucial determinant for hypertension and response to antihypertensive therapy.


Asunto(s)
Hipertensión/metabolismo , Estrés Oxidativo/fisiología , Nexinas de Clasificación/metabolismo , Animales , Presión Sanguínea/fisiología , Línea Celular , Femenino , Humanos , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Transporte de Proteínas/fisiología , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo
2.
Life Sci ; 243: 117226, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31904366

RESUMEN

Hypertension is a risk factor for premature death and roughly 50% of hypertensive patients are salt-sensitive. The incidence of salt-sensitive hypertension increases with age. However, the mechanisms of salt-sensitive hypertension are not well understood. We had demonstrated decreased renal sodium­hydrogen exchanger regulatory factor 1 (NHERF1) expression in old salt-resistant F344 rats. Based on those studies we hypothesized that NHERF1 expression is required for the development of some forms of salt-sensitive hypertension. To address this hypothesis, we measured blood pressure in NHERF1 expressing salt-sensitive 4-mo and 24-mo-old male and female Fischer Brown Norway (FBN) rats male and female 18-mo-old NHERF1 knock-out (NHERF1-/-) mice and wild-type (WT) littermates on C57BL/6J background after feeding high salt (8% NaCl) diet for 7 days. Our data demonstrate that 8% salt diet increased blood pressure in both male and female 24-mo-old FBN rats but not in 4-mo-old FBN rats and in 18-mo-old male and female WT mice but not in NHERF1-/- mice. Renal dopamine 1 receptor (D1R) expression was decreased in 24-mo-old rats, compared with 4-mo-old FBN rats. However, sodium chloride cotransporter (NCC) expression increased in 24-mo-old FBN rats. In FBN rats, age had no effect on NaK ATPase α1 and NKCC2 expression. By contrast, high salt diet increased the renal expressions of NKCC2, and NCC in 24-mo-old FBN rats. High salt diet also increased NKCC2 and NCC expression in WT mice but not NHERF1-/- mice. Our data suggest that renal NHERF1 expression confers salt sensitivity with aging, associated with increased expression of sodium transporters.


Asunto(s)
Envejecimiento/metabolismo , Hipertensión/metabolismo , Fosfoproteínas/fisiología , Cloruro de Sodio Dietético/administración & dosificación , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Femenino , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/genética , Ratas , Ratas Endogámicas F344 , Intercambiadores de Sodio-Hidrógeno/genética
3.
BMC Nephrol ; 20(1): 227, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221135

RESUMEN

BACKGROUND: Cystinuria is an inherited disorder of renal amino acid transport that causes recurrent nephrolithiasis and significant morbidity in humans. It has an incidence of 1 in 7000 worldwide making it one of the most common genetic disorders in man. We phenotypically characterized a mouse model of cystinuria type A resultant from knockout of Slc3a1. METHODS: Knockout of Slc3a1 at RNA and protein levels was evaluated using real-time quantitative PCR and immunofluorescence. Slc3a1 knockout mice were placed on normal or breeder chow diets and evaluated for cystine stone formation over time suing x-ray analysis, and the development of kidney injury by measuring injury biomarkers. Kidney injury was also evaluated via histologic analysis. Amino acid levels were measured in the blood of mice using high performance liquid chromatography. Liver glutathione levels were measured using a luminescent-based assay. RESULTS: We confirmed knockout of Slc3a1 at the RNA level, while Slc7a9 RNA representing the co-transporter was preserved. As expected, we observed bladder stone formation in Slc3a1-/- mice. Male Slc3a1-/- mice exhibited lower weights compared to Slc3a1+/+. Slc3a1-/- mice on a regular diet demonstrated elevated blood urea nitrogen (BUN) without elevation of serum creatinine. However, placing the knockout animals on a breeder chow diet, containing a higher cystine concentration, resulted in the development of elevation of both BUN and creatinine indicative of more severe chronic kidney disease. Histological examination revealed that these dietary effects resulted in worsened kidney tubular obstruction and interstitial inflammation as well as worsened bladder inflammation. Cystine is a precursor for the antioxidant molecule glutathione, so we evaluated glutathione levels in the livers of Slc3a1-/- mice. We found significantly lowered levels of both reduced and total glutathione in the knockout animals. CONCLUSIONS: Our results suggest that that diet can affect the development and progression of chronic kidney disease in an animal model of cystinuria, which may have important implications for patients with this disease. Additionally, reduced glutathione may predispose those with cystinuria to injury caused by oxidative stress. Word count: 327.


Asunto(s)
Nitrógeno de la Urea Sanguínea , Cistinuria/diagnóstico por imagen , Cistinuria/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/deficiencia , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Neutros/deficiencia , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Cistinuria/genética , Femenino , Masculino , Ratones , Ratones Noqueados
4.
FASEB J ; 33(8): 9008-9016, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31042404

RESUMEN

Na+/H+ exchanger regulatory factor 1 (NHERF1; also known as ezrin-radixin-moesin-binding phosphoprotein 50) is a PSD-95, disc large, zona occludens-1 adapter that acts as a scaffold for signaling complexes and cytoskeletal-plasma membrane interactions. NHERF1 is crucial to ß-2-adrenoceptor (ß2AR)-mediated activation of cystic fibrosis transmembrane conductance regulator (CFTR) in epithelial cells, and NHERF1 has been proposed to mediate the recycling of internalized ß2AR back to the cell membrane. In the current study, we assessed the role of NHERF1 in regulating cAMP-mediated signaling and immunomodulatory functions in airway smooth muscle (ASM). NHERF1 knockdown attenuated the induction of (protein kinase A) phospho-vasodilator-stimulated phosphoprotein (p-VASP) by isoproterenol (ISO), prostaglandin E2 (PGE2), or forskolin (FSK) as well as the induction of p-heat shock protein 20 after 4 h of stimulation with ISO and FSK. NHERF1 knockdown fully abrogated the ISO-, PGE2-, and FSK-induced IL-6 gene expression and cytokine production without affecting cAMP-mediated phosphodiesterase 4D (PDE4D) gene expression, phospho-cAMP response element-binding protein (p-CREB), and cAMP response element (CRE)-Luc, or PDGF-induced cyclin D1 expression. Interestingly, NHERF1 knockdown prevented ISO-induced chromatin-binding of the transcription factor CCAAT-enhancer-binding protein-ß (c/EBPß). c/EBPß knockdown almost completely abrogated the cAMP-mediated IL-6 but not PDE4D gene expression. The differential regulation of cAMP-induced signaling and gene expression in our study indicates a role for NHERF1 in the compartmentalization of cAMP signaling in ASM.-Pera, T., Tompkins, E., Katz, M., Wang, B., Deshpande, D. A., Weinman, E. J., Penn, R. B. Specificity of NHERF1 regulation of GPCR signaling and function in human airway smooth muscle.


Asunto(s)
Músculo Liso/metabolismo , Fosfoproteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratorio/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , AMP Cíclico/metabolismo , Ciclina D1/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Músculo Liso/inmunología , Músculo Liso/fisiología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fenómenos Fisiológicos Respiratorios , Sistema Respiratorio/inmunología , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/genética
5.
Am J Physiol Cell Physiol ; 313(2): C197-C206, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515088

RESUMEN

Dopamine decreases Na-K-ATPase (NKA) activity by PKC-dependent phosphorylation and endocytosis of the NKA α1. Dopamine-mediated regulation of NKA is impaired in aging and some forms of hypertension. Using opossum (OK) proximal tubule cells (PTCs), we demonstrated that sodium-hydrogen exchanger regulatory factor-1 (NHERF-1) associates with NKA α1 and dopamine-1 receptor (D1R). This association is required for the dopamine-mediated regulation of NKA. In OK cells, dopamine decreases NHERF-1 association with NKA α1 but increases its association with D1R. However, it is not known whether NHERF-1 plays a role in dopamine-mediated NKA regulation in animal models of hypertension. We hypothesized that defective dopamine-mediated regulation of NKA results from the decrease in NHERF-1 expression in rat renal PTCs isolated from animal models of hypertension [spontaneously hypertensive rats (SHRs) and aged F344 rats]. To test this hypothesis, we isolated and cultured renal PTCs from 22-mo-old F344 rats and their controls, normotensive 4-mo-old F344 rats, and SHRs and their controls, normotensive Wistar-Kyoto (WKY) rats. The results demonstrate that in both hypertensive models (SHR and aged F344), NHERF-1 expression, dopamine-mediated phosphorylation of NKA, and ouabain-inhibitable K+ transport are reduced. Transfection of NHERF-1 into PTCs from aged F344 and SHRs restored dopamine-mediated inhibition of NKA. These results suggest that decreased renal NHERF-1 expression contributes to the impaired dopamine-mediated inhibition of NKA in PTCs from animal models of hypertension.


Asunto(s)
Hipertensión/genética , Túbulos Renales Proximales/metabolismo , Fosfoproteínas/biosíntesis , Intercambiadores de Sodio-Hidrógeno/biosíntesis , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis , Animales , Presión Sanguínea/genética , Línea Celular , Modelos Animales de Enfermedad , Dopamina/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/patología , Masculino , Fosfoproteínas/genética , Ratas , Ratas Endogámicas SHR , Transducción de Señal/genética , Intercambiadores de Sodio-Hidrógeno/genética , ATPasa Intercambiadora de Sodio-Potasio/genética
6.
Neuroscience ; 353: 58-75, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28392297

RESUMEN

The group II metabotropic glutamate receptors mGluR2 and mGluR3 are key modulators of glutamatergic neurotransmission. In order to identify novel Group II metabotropic glutamate receptor (mGluR)-interacting partners, we screened the C-termini of mGluR2 and mGluR3 for interactions with an array of PDZ domains. These screens identified the Na+/H+ exchanger regulatory factors 1 and 2 (NHERF-1 & -2) as candidate interacting partners. Follow-up co-immunoprecipitation studies demonstrated that both mGluR2 and mGluR3 can associate with NHERF-1 and NHERF-2 in a cellular context. Functional studies revealed that disruption of PDZ interactions with mGluR2 enhanced receptor signaling to Akt. However, further studies of mGluR2 and mGluR3 signaling in astrocytes in which NHERF expression was reduced by gene knockout (KO) and/or siRNA knockdown techniques revealed that the observed differences in signaling between WT and mutant mGluR2 were likely not due to disruption of interactions with the NHERF proteins. Electron microscopic analyses revealed that Group II mGluRs were primarily expressed in glia and unmyelinated axons in WT, NHERF-1 and NHERF-2 KO mice, but the relative proportion of labeled axons over glial processes was higher in NHERF-2 KO mice than in controls and NHERF-1 KO mice. Interestingly, our anatomical studies also revealed that loss of either NHERF protein results in ventriculomegaly, which may be related to the high incidence of hydrocephaly that has previously been observed in NHERF-1 KO mice. Together, these studies support a role for NHERF-1 and NHERF-2 in regulating the distribution of Group II mGluRs in the murine brain, while conversely the effects of the mGluR2/3 PDZ-binding motifs on receptor signaling are likely mediated by interactions with other PDZ scaffold proteins beyond the NHERF proteins.


Asunto(s)
Encéfalo/metabolismo , Fosfoproteínas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/ultraestructura , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Nerviosas Amielínicas/metabolismo , Dominios PDZ , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética
7.
J Clin Invest ; 125(9): 3519-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26258413

RESUMEN

Diarrhea is one of the troublesome complications of diabetes, and the underlying causes of this problem are complex. Here, we investigated whether altered electrolyte transport contributes to diabetic diarrhea. We found that the expression of Na+/H+ exchanger NHE3 and several scaffold proteins, including NHE3 regulatory factors (NHERFs), inositol trisphosphate (IP3) receptor-binding protein released with IP3 (IRBIT), and ezrin, was decreased in the intestinal brush border membrane (BBM) of mice with streptozotocin-induced diabetes. Treatment of diabetic mice with insulin restored intestinal NHE3 activity and fluid absorption. Molecular analysis revealed that NHE3, NHERF1, IRBIT, and ezrin form macrocomplexes, which are perturbed under diabetic conditions, and insulin administration reconstituted these macrocomplexes and restored NHE3 expression in the BBM. Silencing of NHERF1 or IRBIT prevented NHE3 trafficking to the BBM and insulin-dependent NHE3 activation. IRBIT facilitated the interaction of NHE3 with NHERF1 via protein kinase D2-dependent phosphorylation. Insulin stimulated ezrin phosphorylation, which enhanced the interaction of ezrin with NHERF1, IRBIT, and NHE3. Additionally, oral administration of lysophosphatidic acid (LPA) increased NHE3 activity and fluid absorption in diabetic mice via an insulin-independent pathway. Together, these findings indicate the importance of NHE3 in diabetic diarrhea and suggest LPA administration as a potential therapeutic strategy for management of diabetic diarrhea.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diarrea/metabolismo , Mucosa Intestinal/metabolismo , Microvellosidades/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Adenosilhomocisteinasa/genética , Adenosilhomocisteinasa/metabolismo , Animales , Células CACO-2 , Complicaciones de la Diabetes/genética , Complicaciones de la Diabetes/patología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diarrea/genética , Diarrea/patología , Humanos , Intestinos/patología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lisofosfolípidos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microvellosidades/genética , Microvellosidades/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética
8.
Hepatology ; 62(4): 1227-36, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26108984

RESUMEN

UNLABELLED: The intercellular adhesion molecule 1 (ICAM-1) is induced in mouse liver after bile duct ligation (BDL) and plays a key role in neutrophil-mediated liver injury in BDL mice. ICAM-1 has been shown to interact with cytoskeletal ezrin-radixin-moesin (ERM) proteins that also interact with the PDZ protein, Na(+) /H(+) exchanger regulatory factor 1 (NHERF-1/EBP50). In NHERF-1(-/-) mice, ERM proteins are significantly reduced in brush-border membranes from kidney and small intestine. ERM knockdown reduces ICAM-1 expression in response to tumor necrosis factor alpha. Here we show that NHERF-1 assembles ERM proteins, ICAM-1 and F-actin into a macromolecule complex that is increased in mouse liver after BDL. Compared to wild-type (WT) mice, both sham-operated and BDL NHERF-1(-/-) mice have lower levels of activated ERM and ICAM-1 protein in the liver accompanied by significantly reduced hepatic neutrophil accumulation, serum alanine aminotransferase, and attenuated liver injury after BDL. However, total bile acid concentrations in serum and liver of sham and BDL NHERF-1(-/-) mice were not significantly different from WT controls, although hepatic tetrahydroxylated bile acids and Cyp3a11 messenger RNA levels were higher in NHERF-1(-/-) BDL mice. CONCLUSION: NHERF-1 participates in the inflammatory response that is associated with BDL-induced liver injury. Deletion of NHERF-1 in mice leads to disruption of the formation of ICAM-1/ERM/NHERF-1 complex and reduction of hepatic ERM proteins and ICAM-1, molecules that are up-regulated and are essential for neutrophil-mediated liver injury in cholestasis. Further study of the role of NHERF-1 in the inflammatory response in cholestasis and other forms of liver injury should lead to discovery of new therapeutic targets in hepatic inflammatory diseases.


Asunto(s)
Colestasis Intrahepática/etiología , Molécula 1 de Adhesión Intercelular/fisiología , Hepatopatías/etiología , Fosfoproteínas/fisiología , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Hepatitis/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromina 2/fisiología , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética
9.
Am J Physiol Renal Physiol ; 309(2): F109-19, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25995109

RESUMEN

Na+/H+ exchanger regulatory factor (NHERF1) plays a critical role in the renal transport of phosphate by binding to Na+-Pi cotransporter (NpT2a) in the proximal tubule. While the association between NpT2a and NHERF1 in the apical membrane is known, the role of NHERF1 to regulate the trafficking of NpT2a has not been studied. To address this question, we performed cell fractionation by sucrose gradient centrifugation in opossum kidney (OK) cells placed in low-Pi medium to stimulate forward trafficking of NpT2a. Immunoblot analysis demonstrated expression of NpT2a and NHERF1 in the endoplasmic reticulum (ER)/Golgi. Coimmunoprecipitation demonstrated a NpT2a-NHERF1 interaction in the ER/Golgi. Low-Pi medium for 4 and 8 h triggered a decrease in NHERF1 in the plasma membrane with a corresponding increase in the ER/Golgi. Time-lapse total internal reflection fluorescence imaging of OK cells placed in low-Pi medium, paired with particle tracking and mean square displacement analysis, indicated active directed movement of NHERF1 at early and late time points, whereas NpT2a showed active movement only at later times. Silence of NHERF1 in OK cells expressing green fluorescent protein (GFP)-NpT2a resulted in an intracellular accumulation of GFP-NpT2a. Transfection with GFP-labeled COOH-terminal (TRL) PDZ-binding motif deleted or wild-type NpT2a in OK cells followed by cell fractionation and immunoprecipitation confirmed that the interaction between NpT2a and NHERF1 was dependent on the TRL motif of NpT2a. We conclude that appropriate trafficking of NpT2a to the plasma membrane is dependent on the initial association between NpT2a and NHERF1 through the COOH-terminal TRL motif of NpT2a in the ER/Golgi and requires redistribution of NHERF1 to the ER/Golgi.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Riñón/metabolismo , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Animales , Línea Celular , Didelphis
10.
J Clin Invest ; 125(5): 2136-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25893600

RESUMEN

Thiazide diuretics are used to treat hypertension; however, compensatory processes in the kidney can limit antihypertensive responses to this class of drugs. Here, we evaluated compensatory pathways in SPAK kinase-deficient mice, which are unable to activate the thiazide-sensitive sodium chloride cotransporter NCC (encoded by Slc12a3). Global transcriptional profiling, combined with biochemical, cell biological, and physiological phenotyping, identified the gene expression signature of the response and revealed how it establishes an adaptive physiology. Salt reabsorption pathways were created by the coordinate induction of a multigene transport system, involving solute carriers (encoded by Slc26a4, Slc4a8, and Slc4a9), carbonic anhydrase isoforms, and V-type H⁺-ATPase subunits in pendrin-positive intercalated cells (PP-ICs) and ENaC subunits in principal cells (PCs). A distal nephron remodeling process and induction of jagged 1/NOTCH signaling, which expands the cortical connecting tubule with PCs and replaces acid-secreting α-ICs with PP-ICs, were partly responsible for the compensation. Salt reabsorption was also activated by induction of an α-ketoglutarate (α-KG) paracrine signaling system. Coordinate regulation of a multigene α-KG synthesis and transport pathway resulted in α-KG secretion into pro-urine, as the α-KG-activated GPCR (Oxgr1) increased on the PP-IC apical surface, allowing paracrine delivery of α-KG to stimulate salt transport. Identification of the integrated compensatory NaCl reabsorption mechanisms provides insight into thiazide diuretic efficacy.


Asunto(s)
Presión Sanguínea/fisiología , Cloruros/orina , Síndrome de Gitelman/fisiopatología , Natriuresis/fisiología , Nefronas/metabolismo , Reabsorción Renal/fisiología , Amilorida/análogos & derivados , Amilorida/farmacología , Amoníaco/metabolismo , Animales , Transporte Biológico , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/fisiología , Modelos Animales de Enfermedad , Activación Enzimática , Canales Epiteliales de Sodio/fisiología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Síndrome de Gitelman/genética , Ácidos Cetoglutáricos/metabolismo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Noqueados , Natriuresis/genética , Comunicación Paracrina , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Notch/fisiología , Receptores Purinérgicos P2/fisiología , Transducción de Señal , Cloruro de Sodio/farmacocinética , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/fisiología , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
11.
Am J Physiol Renal Physiol ; 308(4): F366-76, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25477473

RESUMEN

Sodium-coupled SLC12 cation chloride cotransporters play important roles in cell volume and chloride homeostasis, epithelial fluid secretion, and renal tubular salt reabsorption. These cotransporters are phosphorylated and activated indirectly by With-No-Lysine (WNK) kinases through their downstream effector kinases, Ste20- and SPS1-related proline alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1). Multiple WNK kinases can coexist within a single cell type, although their relative contributions to SPAK/OSR1 activation and salt transport remain incompletely understood. Deletion of specific WNKs from cells that natively express a functional WNK-SPAK/OSR1 network will help resolve these knowledge gaps. Here, we outline a simple method to selectively knock out full-length WNK1 expression from mammalian cells using RNA-guided clustered regularly interspaced short palindromic repeats/Cas9 endonucleases. Two clonal cell lines were generated by using a single-guide RNA (sgRNA) targeting exon 1 of the WNK1 gene, which produced indels that abolished WNK1 protein expression. Both cell lines exhibited reduced endogenous WNK4 protein abundance, indicating that WNK1 is required for WNK4 stability. Consistent with an on-target effect, the reduced WNK4 abundance was associated with increased expression of the KLHL3/cullin-3 E3 ubiquitin ligase complex and was rescued by exogenous WNK1 overexpression. Although the morphology of the knockout cells was indistinguishable from control, they exhibited low baseline SPAK/OSR1 activity and failed to trigger regulatory volume increase after hypertonic stress, confirming an essential role for WNK1 in cell volume regulation. Collectively, our data show how this new, powerful, and accessible gene-editing technology can be used to dissect and analyze WNK signaling networks.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen/métodos , Genoma Humano , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Forma de la Célula , Tamaño de la Célula , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Regulación hacia Abajo , Exones , Genotipo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Microfilamentos , Antígenos de Histocompatibilidad Menor , Presión Osmótica , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Transducción de Señal , Factores de Tiempo , Transfección , Proteína Quinasa Deficiente en Lisina WNK 1
12.
Nat Commun ; 5: 5482, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25405894

RESUMEN

Primary cilia contain specific receptors and channel proteins that sense the extracellular milieu. Defective ciliary function causes ciliopathies such as autosomal dominant polycystic kidney disease (ADPKD). However, little is known about how large ciliary transmembrane proteins traffic to the cilia. Polycystin-1 (PC1) and -2 (PC2), the two ADPKD gene products, are large transmembrane proteins that co-localize to cilia where they act to control proper tubular diameter. Here we describe that PC1 and PC2 must interact and form a complex to reach the trans-Golgi network (TGN) for subsequent ciliary targeting. PC1 must also be proteolytically cleaved at a GPS site for this to occur. Using yeast two-hybrid screening coupled with a candidate approach, we identify a Rabep1/GGA1/Arl3-dependent ciliary targeting mechanism, whereby Rabep1 couples the polycystin complex to a GGA1/Arl3-based ciliary trafficking module at the TGN. This study provides novel insights into the ciliary trafficking mechanism of membrane proteins.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Cilios/metabolismo , Canales Catiónicos TRPP/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Red trans-Golgi/metabolismo , Factores de Ribosilacion-ADP/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Cilios/genética , Riñón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Transporte de Proteínas , Canales Catiónicos TRPP/genética , Proteínas de Transporte Vesicular/genética , Red trans-Golgi/genética
13.
Hypertension ; 64(4): 825-32, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25024286

RESUMEN

The dopamine D2 receptor (D2R) decreases renal reactive oxygen species (ROS) production and regulates blood pressure, in part, via positive regulation of paraoxonase 2. Sestrin2, a highly conserved antioxidant protein, regulates intracellular ROS level by regenerating hyperoxidized peroxiredoxins. We hypothesized that sestrin2 may be involved in preventing excessive renal ROS production and thus contribute to the maintenance of normal blood pressure. Moreover, the D2R may decrease ROS production, in part, through the regulation of sestrin2. Renal sestrin2 expression was lower (-62±13%) in D2R(-/-) than in D2R(+/+) mice. Silencing D2R in human renal proximal tubule cells decreased sestrin2 expression (-53±3%) and increased hyperoxidized peroxiredoxins (2.9-fold). Stimulation of D2R in renal proximal tubule cells increased sestrin2 expression (1.6-fold), decreased hyperoxidized peroxiredoxins (-61±3%), and reduced ROS production (-31±4%). Silencing sestrin2 in renal proximal tubule cells increased hyperoxidized peroxiredoxins (2.1-fold) and ROS production (1.3-fold). Silencing sestrin2 also abolished D2R-induced decrease in peroxiredoxin hyperoxidation and partially prevented the inhibitory effect of D2R stimulation on ROS production. Silencing paraoxonase 2 increased sestrin2 ubiquitinylation (2.8-fold), decreased sestrin2 expression (-30±3%), and increased ROS production (1.3-fold), peroxiredoxin hyperoxidation (2.9-fold), and lipid peroxidation (2.3-fold), and blocked the increase in sestrin2 that occurs with D2R stimulation. In vivo renal selective silencing of sestrin2 by the renal subcapsular infusion of sestrin2 small interfering RNA (3 µg/day; 7 days) in mice increased renal oxidative stress (1.3-fold) and blood pressure. These results suggest that the D2R, via paraoxonase 2 and sestrin2, keeps normal renal redox balance, which contributes to the maintenance of normal blood pressure.


Asunto(s)
Presión Sanguínea/fisiología , Riñón/metabolismo , Proteínas Nucleares/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Células Cultivadas , Agonistas de Dopamina/farmacología , Humanos , Immunoblotting , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Oxidación-Reducción , Peroxidasas , Peroxirredoxinas/metabolismo , Quinpirol/farmacología , Interferencia de ARN , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/genética , Regulación hacia Arriba/efectos de los fármacos
14.
Cell Signal ; 26(11): 2521-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25049074

RESUMEN

Dopamine D1-like receptors (D1R and D5R) stimulate adenylyl cyclase (AC) activity, whereas the D2-like receptors (D2, D3 and D4) inhibit AC activity. D1R, but not the D5R, has been reported to regulate AC activity in lipid rafts (LRs). We tested the hypothesis that D1R and D5R differentially regulate AC activity in LRs using human embryonic kidney (HEK) 293 cells heterologously expressing human D1 or D5 receptor (HEK-hD1R or HEK-hD5R) and human renal proximal tubule (hRPT) cells that endogenously express D1R and D5R. Of the AC isoforms expressed in HEK and hRPT cells (AC3, AC5, AC6, AC7, and AC9), AC5/6 was distributed to a greater extent in LRs than non-LRs in HEK-hD1R (84.5±2.3% of total), HEK-hD5R (68.9±3.1% of total), and hRPT cells (66.6 ± 2.2% of total) (P<0.05, n=4/group). In HEK-hD1R cells, the D1-like receptor agonist fenoldopam (1 µM/15 min) increased AC5/6 protein (+17.2 ± 3.9% of control) in LRs but decreased it in non-LRs (-47.3±5.3% of control) (P<0.05, vs. control, n=4/group). By contrast, in HEK-hD5R cells, fenoldopam increased AC5/6 protein in non-LRs (+67.1 ± 5.3% of control, P<0.006, vs. control, n=4) but had no effect in LRs. In hRPT cells, fenoldopam increased AC5/6 in LRs but had little effect in non-LRs. Disruption of LRs with methyl-ß-cyclodextrin decreased basal AC activity in HEK-D1R (-94.5 ± 2.0% of control) and HEK-D5R cells (-87.1 ± 4.6% of control) but increased it in hRPT cells (6.8±0.5-fold). AC6 activity was stimulated to a greater extent by D1R than D5R, in agreement with the greater colocalization of AC5/6 with D1R than D5R in LRs. We conclude that LRs are essential not only for the proper membrane distribution and maintenance of AC5/6 activity but also for the regulation of D1R- and D5R-mediated AC signaling.


Asunto(s)
Adenilil Ciclasas/metabolismo , Túbulos Renales Proximales/metabolismo , Microdominios de Membrana/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D5/metabolismo , Transducción de Señal/fisiología , Adenilil Ciclasas/genética , Línea Celular , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Túbulos Renales Proximales/citología , Microdominios de Membrana/genética , Receptores de Dopamina D1/genética , Receptores de Dopamina D5/genética
15.
Am J Kidney Dis ; 62(5): 1006-11, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23769135

RESUMEN

Increases in serum concentrations of parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) and ultimately phosphate and decreases in 1,25-dihydroxyvitamin D level are thought to play a central role in the progressive nature of kidney disease and the development of cardiovascular disease in patients with chronic kidney disease. The initial changes in PTH and FGF-23 levels are adaptive to maintain serum phosphate concentration and phosphate load within defined levels by increasing urinary excretion of phosphate. Less well appreciated is the unanticipated finding that absorption of phosphate from the gastrointestinal tract is not downregulated in chronic kidney disease. This maladaptive response maintains higher levels of phosphate absorption, thereby contributing to the phosphate burden. Moreover, in response to a low-phosphate diet, as often is prescribed to such patients, gut phosphate absorption may be enhanced, undermining the potential beneficial effects of this intervention. Given the poor response to limiting phosphate intake and the use of phosphate binders, we suggest that research efforts be oriented toward better understanding of the factors that affect phosphate absorption in the gastrointestinal tract and the development of agents that directly inhibit phosphate transporters in the small intestine and/or their associated binding proteins.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Tracto Gastrointestinal/metabolismo , Hiperfosfatemia/metabolismo , Fosfatos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Adulto , Enfermedades Cardiovasculares/metabolismo , Dieta , Progresión de la Enfermedad , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hiperfosfatemia/complicaciones , Hiperfosfatemia/prevención & control , Masculino , Cooperación del Paciente , Proteínas de Unión a Fosfato/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Factores de Riesgo
16.
Hypertens Res ; 36(8): 684-90, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23425954

RESUMEN

D5 dopamine receptor (D5R) knock-out mice (D5(-/-)) have a higher blood pressure (BP) and higher reactive oxygen species (ROS) production than their D5R wild-type littermates (D5(+/+)). We tested the hypothesis that the high BP and increased ROS production in D5(-/-) mice may be caused by decreased heme oxygenase-1 (HO-1) expression and activity. We found that renal HO-1 protein expression and HO enzyme activity were decreased (65 and 50%, respectively) in D5(-/-) relative to D5(+/+) mice. A 24 h of administration of hemin, an HO-1 inducer, increased HO-1 expression and HO activity (6.8- and 1.9-fold, respectively) and normalized the increased ROS production and BP in D5(-/-) mice. Expression of HO-1 protein and HO activity were increased (2.3- and 1.5-fold, respectively) in HEK cells that heterologously expressed human wild-type D5R (HEK-hD5R), but not the empty vector-transfected HEK-293 cells. Fenoldopam (Fen), a D5R agonist, increased HO activity (3 h), HO-1 protein expression, HO-1 and D5R colocalization and co-immunoprecipitation in HEK-hD5R cells. Cellular NADPH oxidase activity was decreased by 35% in HEK-hD5R that was abrogated with silencing of the heme oxygenase 1 gene (HMOX1). HMOX1 siRNA also impaired the ability of Fen to decrease NADPH oxidase activity in HEK-hD5R cells. In summary, the D5R positively regulates HO-1 through direct protein/protein interaction in the short-term and by increasing HO-1 protein expression in the long-term. The impaired D5R regulation of HO-1 and ROS production contributes to the pathogenesis of hypertension in D5(-/-) mice.


Asunto(s)
Presión Sanguínea/fisiología , Hemo-Oxigenasa 1/metabolismo , Hipertensión/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Dopamina D5/metabolismo , Animales , Agonistas de Dopamina/farmacología , Fenoldopam/farmacología , Células HEK293 , Humanos , Hipertensión/enzimología , Hipertensión/genética , Ratones , Ratones Noqueados , Receptores de Dopamina D5/genética , Regulación hacia Arriba/efectos de los fármacos
17.
Clin Nephrol ; 80(6): 464-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22784561

RESUMEN

While the classic electrocardiographic (EKG) findings of hyperkalemia are well known to clinicians, the association between hyperkalemia and bradycardia is not widely appreciated. Three cases of profound bradycardia due to hyperkalemia in patients with End Stage Renal Disease (ESRD) on hemodialysis are described to provide a base for discussion of specific issues in the management of such patients. The patients presented with hyperkalemia and severe bradycardia that did not improve after administration of atropine. Urgent hemodialysis in two cases led to resolution of the bradycardia. In the third case, the failure to recognize that bradycardia was the consequence of the hyperkalemia led to unnecessary interventions and delays in initiating dialysis. These cases highlight the causal relation between hyperkalemia and bradycardia in ESRD patients and emphasize the need for increased awareness of this association.


Asunto(s)
Bradicardia/etiología , Electrocardiografía , Hiperpotasemia/complicaciones , Diálisis Renal/efectos adversos , Humanos , Fallo Renal Crónico/complicaciones , Masculino , Persona de Mediana Edad
18.
J Biol Chem ; 287(42): 35047-35056, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22904329

RESUMEN

P(i) uptake in the small intestine occurs predominantly through the NaPi-2b (SLC34a2) co-transporter. NaPi-2b is regulated by changes in dietary P(i) but the mechanisms underlying this regulation are largely undetermined. Sequence analyses show NaPi-2b has a PDZ binding motif at its C terminus. Immunofluorescence imaging shows NaPi-2b and two PDZ domain containing proteins, NHERF1 and PDZK1, are expressed in the apical microvillar domain of rat small intestine enterocytes. Co-immunoprecipitation studies in rat enterocytes show that NHERF1 associates with NaPi-2b but not PDZK1. In HEK co-expression studies, GFP-NaPi-2b co-precipitates with FLAG-NHERF1. This interaction is markedly diminished when the C-terminal four amino acids are truncated from NaPi-2b. FLIM-FRET analyses using tagged proteins in CACO-2(BBE) cells show a distinct phasor shift between NaPi-2b and NHERF1 but not between NaPi-2b and the PDZK1 pair. This shift demonstrates that NaPi-2b and NHERF1 reside within 10 nm of each other. NHERF1(-/-) mice, but not PDZK1(-/-) mice, had a diminished adaptation of NaPi-2b expression in response to a low P(i) diet. Together these studies demonstrate that NHERF1 associates with NaPi-2b in enterocytes and regulates NaPi-2b adaptation.


Asunto(s)
Enterocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/biosíntesis , Animales , Células CACO-2 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto , Enterocitos/citología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Noqueados , Microvellosidades/genética , Microvellosidades/metabolismo , Fosfoproteínas/genética , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Intercambiadores de Sodio-Hidrógeno/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/genética
19.
Mol Biol Cell ; 23(11): 2028-40, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22496422

RESUMEN

Metastatic cells are highly plastic for differential expression of tumor phenotype hallmarks and metastatic organotropism. The signaling proteins orchestrating the shift of one cell phenotype and organ pattern to another are little known. Na(+)/H(+) exchanger regulatory factor (NHERF1) is a molecular pathway organizer, PDZ-domain protein that recruits membrane, cytoplasmic, and cytoskeletal signaling proteins into functional complexes. To gain insight into the role of NHERF1 in metastatic progression, we stably transfected a metastatic breast cell line, MDA-MB-231, with an empty vector, with wild-type NHERF1, or with NHERF1 mutated in either the PDZ1- or PDZ2-binding domains to block their binding activities. We observed that NHERF1 differentially regulates the expression of two phenotypic programs through its PDZ domains, and these programs form the mechanistic basis for metastatic organotropism. The PDZ2 domain promotes visceral metastases via increased invadopodia-dependent invasion and anchorage-independent growth, as well as by inhibition of apoptosis, whereas the PDZ1 domain promotes bone metastases by stimulating podosome nucleation, motility, neoangiogenesis, vasculogenic mimicry, and osteoclastogenesis in the absence of increased growth or invasion. Collectively, these findings identify NHERF1 as an important signaling nexus for coordinating cell structure with metastatic behavior and identifies the "mesenchymal-to-vasculogenic" phenotypic transition as an essential step in metastatic progression.


Asunto(s)
Dominios PDZ , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo , Tropismo , Animales , Apoptosis , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Matriz Extracelular/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Seudópodos/metabolismo , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Am J Physiol Renal Physiol ; 303(3): F321-7, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22535796

RESUMEN

The renal excretion of inorganic phosphate is regulated in large measure by three hormones, namely, parathyroid hormone, dopamine, and fibroblast growth factor-23. Recent experiments have indicated that the major sodium-dependent phosphate transporter in the renal proximal tubule, Npt2a, binds to the adaptor protein sodium-hydrogen exchanger regulatory factor-1 (NHERF-1) and in the absence of NHERF-1, the inhibitory effect of these three hormones is absent. From these observations, a new model for the hormonal regulation of renal phosphate transport was developed. The downstream signaling pathways of these hormones results in the phosphorylation of the PDZ 1 domain of NHERF-1 and the dissociation of Npt2a/NHERF-1 complexes. In turn, this dissociation facilitates the endocytosis of Npt2a with a subsequent decrease in the apical membrane abundance of the transporter and a decrease in phosphate reabsorption. The current review outlines the experimental observations supporting the operation of this unique regulatory system.


Asunto(s)
Dopamina/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Hormonas/metabolismo , Riñón/metabolismo , Hormona Paratiroidea/fisiología , Fosfatos/metabolismo , Fosfoproteínas/fisiología , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Transporte Biológico Activo/fisiología , Factor-23 de Crecimiento de Fibroblastos , Humanos , Modelos Moleculares , Dominios PDZ/fisiología , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación , Conformación Proteica , Transducción de Señal/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...