Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 399
Filtrar
Más filtros












Intervalo de año de publicación
1.
Sci Rep ; 12(1): 496, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017633

RESUMEN

The in vivo function of cell-derived extracellular vesicles (EVs) is challenging to establish since cell-specific EVs are difficult to isolate and differentiate. We, therefore, created an EV reporter using truncated CD9 to display enhanced green fluorescent protein (EGFP) on the EV surface. CD9truc-EGFP expression in cells did not affect EV size and concentration but enabled co-precipitation of EV markers TSG101 and ALIX from the cell-conditioned medium by anti-GFP immunoprecipitation. We then created a transgenic mouse where CD9truc-EGFP was inserted in the inverse orientation and double-floxed, ensuring irreversible Cre recombinase-dependent EV reporter expression. We crossed the EV reporter mice with mice expressing Cre ubiquitously (CMV-Cre), in cardiomyocytes (αMHC-MerCreMer) and renal tubular epithelial cells (Pax8-Cre), respectively. The CD9truc-EGFP positive mice showed Cre-dependent EGFP expression, and plasma CD9truc-EGFP EVs were immunoprecipitated only from CD9truc-EGFP positive CD9truc-EGFPxCMV-Cre and CD9truc-EGFPxαMHC-Cre mice, but not in CD9truc-EGFPxPax8-Cre and CD9truc-EGFP negative mice. In urine samples, CD9truc-EGFP EVs were detected by immunoprecipitation only in CD9truc-EGFP positive CD9truc-EGFPxCMV-Cre and CD9truc-EGFPxPax8-Cre mice, but not CD9truc-EGFPxαMHC-Cre and CD9truc-EGFP negative mice. In conclusion, our EV reporter mouse model enables Cre-dependent EV labeling, providing a new approach to studying cell-specific EVs in vivo and gaining a unique insight into their physiological and pathophysiological function.


Asunto(s)
Vesículas Extracelulares/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones Transgénicos/genética , Animales , Células Epiteliales/metabolismo , Vesículas Extracelulares/química , Vesículas Extracelulares/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Ratones , Ratones Transgénicos/metabolismo , Miocitos Cardíacos/metabolismo , Especificidad de Órganos , Transgenes
2.
Sci Rep ; 11(1): 8280, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859322

RESUMEN

Severe renal ischemia-reperfusion injury (IRI) can lead to acute and chronic kidney dysfunction. Cytoskeletal modifications are among the main effects of this condition. The majority of studies that have contributed to the current understanding of IRI have relied on histological analyses using exogenous probes after the fact. Here we report the successful real-time visualization of actin cytoskeletal alterations in live proximal and distal tubules that arise at the onset of severe IRI. To achieve this, we induced fluorescent actin expression in these segments in rats with hydrodynamic gene delivery (HGD). Using intravital two-photon microscopy we then tracked and quantified endogenous actin dysregulation that occurred by subjecting these animals to 60 min of bilateral renal ischemia. Rapid (by 1-h post-reperfusion) and significant (up to 50%) declines in actin content were observed. The decline in fluorescence within proximal tubules was significantly greater than that observed in distal tubules. Actin-based fluorescence was not recovered during the measurement period extending 24 h post-reperfusion. Such injury decimated the renal architecture, in particular, actin brush borders, and hampered the reabsorptive and filtrative capacities of these tubular compartments. Thus, for the first time, we show that the combination of HGD and intravital microscopy can serve as an experimental tool to better understand how IRI modifies the cytoskeleton in vivo and provide an extension to current histopathological techniques.


Asunto(s)
Actinas/metabolismo , Isquemia/diagnóstico por imagen , Isquemia/metabolismo , Túbulos Renales Distales/diagnóstico por imagen , Túbulos Renales Distales/metabolismo , Túbulos Renales Proximales/diagnóstico por imagen , Túbulos Renales Proximales/metabolismo , Riñón/irrigación sanguínea , Imagen Molecular/métodos , Daño por Reperfusión/diagnóstico por imagen , Daño por Reperfusión/metabolismo , Animales , Citoesqueleto/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Proximales/citología , Ratas , Índice de Severidad de la Enfermedad
3.
Acta Histochem ; 123(4): 151701, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33691202

RESUMEN

Pre-clinical animal models are needed to investigate and study kidney injuries and diseases. The rabbit kidney model is frequently used because various important parameters can be assessed with it. For example, histology and immunohistochemistry are indispensable as tissue morphology and composition can be investigated qualitatively as well as quantitatively. Here, different histological and immunohistochemical stainings were performed in the rabbit healthy naïve kidney tissue. First, overnight formalin fixation followed by paraffin embedding and cryopreservation with a subsequent 10-minute formalin fixation prior to staining were compared. Cryosections showed a more pronounced staining pattern, with clear borders at low magnifications, but blurred borders at higher magnifications. Then, antigen retrieval (AR) for paraffin embedded sections resulted in more prominent corresponding signals compared to stainings without AR. Moreover, several advantages and disadvantages of chromogenic versus immunofluorescence stainings were considered. Chromogenic staining was advantageous compared to immunofluorescence for collagen I and III, and to a minor degree for fibronectin. Finally, distinct structures, such as the pelvis, the calices, the glomeruli and tubuli, were stained in serial sections with diverse immunohistochemical stainings in order to delineate their composition. The following stainings were performed: standard Haematoxylin&Eosin and Elastica van Gieson staining, collagen I, collagen III, fibronectin, α-SMA, ki-67 and protease-activated receptor-2 (PAR-2). While chromogenic stainings of collagen I and collagen III were particularly useful to depict kidney structures in paraffin sections compared with cryosections, cryosections immunofluorescently stained for α-SMA were superior to paraffin sections, particularly at higher magnifications. With regard to specific structures, we found renal vessel walls positive for fibronectin and α-SMA, while the Bowman's capsule was only positive for fibronectin and α-SMA showed only tiny spots. The mesangial cells of the glomeruli and the distal tubuli were PAR-2 positive, while the proximal tubuli were PAR-2 negative.


Asunto(s)
Inmunohistoquímica , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Células Mesangiales/citología , Células Mesangiales/metabolismo , Coloración y Etiquetado , Animales , Femenino , Adhesión en Parafina , Conejos
4.
Hypertension ; 77(3): 868-881, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33486984

RESUMEN

Genome-wide association studies have identified that NPR-C (natriuretic peptide receptor-C) variants are associated with elevation of blood pressure. However, the mechanism underlying the relationship between NPR-C and blood pressure regulation remains elusive. Here, we investigate whether NPR-C regulates Ang II (angiotensin II)-induced hypertension through sodium transporters activity. Wild-type mice responded to continuous Ang II infusion with an increased renal NPR-C expression. Global NPR-C deficiency attenuated Ang II-induced increased blood pressure both in male and female mice associated with more diuretic and natriuretic responses to a saline challenge. Interestingly, Ang II increased both total and phosphorylation of NCC (NaCl cotransporter) abundance involving in activation of WNK4 (with-no-lysine kinase 4)/SPAK (Ste20-related proline/alanine-rich kinase) which was blunted by NPR-C deletion. NCC inhibitor, hydrochlorothiazide, failed to induce natriuresis in NPR-C knockout mice. Moreover, low-salt and high-salt diets-induced changes of total and phosphorylation of NCC expression were normalized by NPR-C deletion. Importantly, tubule-specific deletion of NPR-C also attenuated Ang II-induced elevated blood pressure, total and phosphorylation of NCC expression. Mechanistically, in distal convoluted tubule cells, Ang II dose and time-dependently upregulated WNK4/SPAK/NCC kinase pathway and NPR-C/Gi/PLC/PKC signaling pathway mediated NCC activation. These results demonstrate that NPR-C signaling regulates NCC function contributing to sodium retention-mediated elevated blood pressure, which suggests that NPR-C is a promising candidate for the treatment of sodium retention-related hypertension.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Riñón/metabolismo , Receptores del Factor Natriurético Atrial/deficiencia , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Angiotensina II , Animales , Presión Sanguínea/genética , Células Cultivadas , Femenino , Hipertensión/inducido químicamente , Hipertensión/genética , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores del Factor Natriurético Atrial/genética , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/genética , Sodio/sangre , Sodio/orina , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
5.
Pflugers Arch ; 473(1): 79-93, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33200256

RESUMEN

The renal distal convoluted tubule (DCT) is critical for the fine-tuning of urinary ion excretion and the control of blood pressure. Ion transport along the DCT is tightly controlled by posttranscriptional mechanisms including a complex interplay of kinases, phosphatases, and ubiquitin ligases. Previous work identified the transcription factor Prox-1 as a gene significantly enriched in the DCT of adult mice. To test if Prox-1 contributes to the transcriptional regulation of DCT function and structure, we developed a novel mouse model (NCCcre:Prox-1flox/flox) for an inducible deletion of Prox-1 specifically in the DCT. The deletion of Prox-1 had no obvious impact on DCT structure and growth independent whether the deletion was achieved in newborn or adult mice. Furthermore, DCT-specific Prox-1 deficiency did not alter DCT-proliferation in response to loop diuretic treatment. Likewise, the DCT-specific deletion of Prox-1 did not cause other gross phenotypic abnormalities. Body weight, urinary volume, Na+ and K+ excretion as well as plasma Na+, K+, and aldosterone levels were similar in Prox-1DCTKO and Prox-1DCTCtrl mice. However, Prox-1DCTKO mice exhibited a significant hypomagnesemia with a profound downregulation of the DCT-specific apical Mg2+ channel TRPM6 and the NaCl cotransporter (NCC) at both mRNA and protein levels. The expression of other proteins involved in distal tubule Mg2+ and Na+ handling was not affected. Thus, Prox-1 is a DCT-enriched transcription factor that does not control DCT growth but contributes to the molecular control of DCT-dependent Mg2+ homeostasis in the adult kidney.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Túbulos Renales Distales/efectos de los fármacos , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Canales Catiónicos TRPM/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Eliminación de Gen , Proteínas de Homeodominio/genética , Túbulos Renales Distales/citología , Magnesio/metabolismo , Ratones , Potasio/metabolismo , Sodio/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Canales Catiónicos TRPM/genética , Proteínas Supresoras de Tumor/genética
6.
Mol Med Rep ; 22(4): 3008-3016, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32945396

RESUMEN

Angiotensin­converting enzyme 2 (ACE2), an important component of the renin­angiotensin system, protects against renal tubulointerstitial fibrosis, but its level of involvement in the mechanism of diabetic nephropathy (DN) currently remains unclear. Herein, the effects of ACE2 in DN and the associated mechanisms were investigated using serum and renal biopsy specimens from patients with DN and control participants, and human renal proximal tubular epithelial cells (HRPTEpiCs). The present study determined that the circulating concentration of ACE2 was high, but renal ACE2 expression was markedly lower, and there was abundant expression of Arkadia, an E3 ubiquitin ligase, in patients with DN. In vitro, ACE2 attenuated high­glucose­induced tubular epithelial to mesenchymal cell transition (EMT), which was demonstrated by increased expression of α­SMA and loss of E­cadherin expression, as demonstrated by western blot analysis and reverse transcription­quantitative PCR. Adenovirus­mediated ACE2 overexpression was also revealed to significantly inhibit Arkadia expression and alleviated high­glucose­induced EMT, while ACE2 inhibition had the opposite effects. Furthermore, western blot analysis demonstrated that ACE2­alleviated EMT was associated with downregulated Arkadia and increased SMAD family member 7 (Smad7) protein, followed by TGF­ß/Smad pathway inhibition in HRPTEpiCs. In conclusion, ACE2 is protective in DN, which may be due to the inhibition of Arkadia­mediated Smad7 degradation, whereby TGF­ß/Smad­mediated EMT is ameliorated in high­glucose­stimulated HRPTEpiCs.


Asunto(s)
Enzima Convertidora de Angiotensina 2/sangre , Nefropatías Diabéticas/metabolismo , Glucosa/efectos adversos , Túbulos Renales Distales/citología , Proteína smad7/metabolismo , Regulación hacia Arriba , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/farmacología , Animales , Estudios de Casos y Controles , Línea Celular , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Riñón/metabolismo , Túbulos Renales Distales/efectos de los fármacos , Túbulos Renales Distales/metabolismo , Masculino , Persona de Mediana Edad , Proteína smad7/genética
7.
Kidney Int ; 97(1): 119-129, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31685313

RESUMEN

Tubulointerstitial disease in the kidney culminates in renal fibrosis that portents organ failure. Twist1, a basic helix-loop-helix protein 38 transcription factor, regulates several essential biological functions, but inappropriate Twist1 activity in the kidney epithelium can trigger kidney fibrogenesis and chronic kidney disease. By contrast, Twist1 in circulating myeloid cells may constrain inflammatory injury by attenuating cytokine generation. To dissect the effects of Twist1 in kidney tubular versus immune cells on renal inflammation following toxin-induced renal injury, we subjected mice with selective deletion of Twist1 in renal epithelial cells or macrophages to aristolochic acid-induced chronic kidney disease. Ablation of Twist1 in the distal nephron attenuated kidney damage, interstitial fibrosis, and renal inflammation after aristolochic acid exposure. However, macrophage-specific deletion of Twist1 did not impact the development of aristolochic acid-induced nephropathy. In vitro studies confirmed that Twist1 in renal tubular cells underpins their susceptibility to apoptosis and propensity to generate pro-fibrotic mediators in response to aristolochic acid. Moreover, co-culture studies revealed that Twist1 in renal epithelia augmented the recruitment and activation of pro-inflammatory CD64+ macrophages. Thus, Twist1 in the distal nephron rather than in infiltrating macrophages propagates chronic inflammation and fibrogenesis during aristolochic acid-induced nephropathy.


Asunto(s)
Túbulos Renales Distales/patología , Macrófagos/inmunología , Nefritis Intersticial/inmunología , Insuficiencia Renal Crónica/inmunología , Proteína 1 Relacionada con Twist/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Ácidos Aristolóquicos/toxicidad , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Túbulos Renales Distales/citología , Túbulos Renales Distales/inmunología , Túbulos Renales Distales/metabolismo , Lipocalina 2/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Nefritis Intersticial/inducido químicamente , Nefritis Intersticial/patología , Cultivo Primario de Células , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/patología , Proteína 1 Relacionada con Twist/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
8.
Physiol Rep ; 7(22): e14280, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31762176

RESUMEN

A potassium (K+ ) rich diet is known to have an antihypertensive effect that has been embodied by the NHLBI in the DASH diet. However, the molecular basis for this blood pressure-lowering effect has been unclear, until a recent study proposed a model in which the DCT cells of the kidney regulate their salt transport in response to variations in intracellular chloride ([Cl- ]i ), which are directly regulated by serum K+ . With the knowledge that WNK proteins are Cl- sensors, and are a part of the WNK/SPAK/NCC signaling cascade which regulates the NCC, the main salt transporter in the distal nephron, we examined the effect of serum K+ on the ([Cl- ]i ) and, in turn its effect on the WNK4 signaling pathway in a "modified HEK 293T" cell line. Using a fluorescence-based approach in this cell line, we have shown that the membrane potential of the cell membrane is sensitive to the small changes in external KCl within the physiological range (2-5 mM), thus functioning as a K+ electrode. When the extracellular K+ was progressively increased (2-5 mM), the membrane depolarization lead to a subsequent increase in [Cl- ]i measured by fluorescence quenching of an intracellular chloride sensor. Increase in extracellular [K] resulted in a decrease in the phosphorylation of the WNK4 protein and its downstream targets, SPAK and NCC. This confirms that small changes in serum K can affect WNK4/SPAK/NCC signaling and transcellular Na+ flux through the DCT and provide a possible mechanism by which a K-rich DASH diet could reduce blood pressure.


Asunto(s)
Líquido Extracelular/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Cloruro de Potasio/metabolismo , Cloruro de Potasio/farmacología , Animales , Relación Dosis-Respuesta a Droga , Líquido Extracelular/efectos de los fármacos , Células HEK293 , Humanos , Túbulos Renales Distales/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones
9.
J Am Soc Nephrol ; 30(5): 782-794, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30914436

RESUMEN

BACKGROUND: Mechanisms underlying the frequent association between salt-sensitive hypertension and type 2 diabetes remain obscure. We previously found that protein kinase C (PKC) activation phosphorylates Kelch-like 3 (KLHL3), an E3 ubiquitin ligase component, at serine 433. We investigated whether impaired KLHL3 activity results in increased renal salt reabsorption via NaCl cotransporter (NCC). METHODS: We used the db/db diabetes mouse model to explore KLHL3's role in renal salt handling in type 2 diabetes and evaluated mechanisms of KLHL3 dysregulation in cultured cells. RESULTS: We observed PKC activity in the db/db mouse kidney and phosphorylation of serine 433 in KLHL3 (KLHL3S433-P). This modification prevents binding of with-no-lysine (WNK) kinases; however, total KLHL3 levels were decreased, indicating severely impaired KLHL3 activity. This resulted in WNK accumulation, activating NCC in distal convoluted tubules. Ipragliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, lowered PKC activity in distal convoluted tubule cells and reduced KLHL3S433-P and NCC levels, whereas the thiazolidinedione pioglitazone did not, although the two agents similarly reduced in blood glucose levels. We found that, in human embryonic kidney cells expressing KLHL3 and distal convoluted tubule cells, cellular glucose accumulation increased KLHL3S433-P levels through PKC. Finally, the effect of PKC inhibition in the kidney of db/db mice confirmed PKC's causal role in KLHL3S433-P and NCC induction. CONCLUSIONS: Dysregulation of KLHL3 is involved in the pathophysiology of type 2 diabetes. These data offer a rationale for use of thiazide in individuals with diabetes and provide insights into the mechanism for cardiorenal protective effects of SGLT2 inhibitors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Glucósidos/farmacología , Proteínas de Microfilamentos/genética , Proteína Quinasa C/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Tiofenos/farmacología , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Túbulos Renales Distales/citología , Ratones , Ratones Obesos , Proteínas de Microfilamentos/metabolismo , Fosforilación , Sensibilidad y Especificidad , Transducción de Señal
10.
Biomed Pharmacother ; 99: 956-969, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29710496

RESUMEN

Dehydrocostus lactone (DHCL), a sesquiterpene lactone is well-known for its antiulcer, anti-hepatotoxic and anticancer activity. However, the studies concerning the safety/toxicity potential of DHCL toward the cells of normal origin remain unclear. The present study is aimed at investigating the toxicity potential of DHCL in renal distal tubular and interstitial fibroblast cell lines (MDCK and NRK-49F cells, respectively), and also in ovarian epithelial cell line (CHO cells). The MTT assay has predicted potential cytotoxic activity of DHCL against the cell line types with IC50 values of 0.99, 2.1 and 5.15??M, respectively. The prominent dose-dependent (IC30,50 & 70) increase in the percentage of cells at subG1 phase in all the cell lines revealed apoptosis induction, further establishing the cytotoxic effect of DHCL. The DHCL exposure (4?h) revealed the induction of ROS in both renal cell lines, which is responsible for apoptosis induction. The NRK-49F cells displayed dose-wise (IC30-70) increase in chromatin condensation and membranous phosphatidylserine translocation further confirming apoptotic cell death. Also, their increase in BAX/Bcl-2 ratio, mitochondrial membrane permeability and caspase-3/7 activity establishes mitochondrial mediated apoptosis. In case of CHO cells, the higher percentage of cells at G2/M phase and expression of Cyclin B1 at lower concentration of DHCL (?IC30), indicate mitotic arrest. The incidence of chromatid gaps and negligible micronuclei formation in treated cells (IC10-30) suggest that sub-lethal concentrations of DHCL exposure causes mitotic arrest in response to the damages by steady expression of Cyclin B1. Under in vitro condition, the study of DHCL's potential cytotoxic effect on both kidney cells and ovarian epithelial cells indicated the possibility of adverse effects on normal healthy cells as well. Hence, the study recommends in-depth investigations on DHCL usage concerning its safety in therapeutic applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Lactonas/farmacología , Sesquiterpenos/farmacología , Animales , Células CHO , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Cricetinae , Cricetulus , Perros , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Femenino , Fibroblastos/citología , Concentración 50 Inhibidora , Túbulos Renales Distales/citología , Túbulos Renales Distales/efectos de los fármacos , Lactonas/administración & dosificación , Células de Riñón Canino Madin Darby , Ovario/citología , Ovario/efectos de los fármacos , Ratas , Sesquiterpenos/administración & dosificación
11.
Angiogenesis ; 21(3): 617-634, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29627966

RESUMEN

The kidney vasculature facilitates the excretion of wastes, the dissemination of hormones, and the regulation of blood chemistry. To carry out these diverse functions, the vasculature is regionalized within the kidney and along the nephron. However, when and how endothelial regionalization occurs remains unknown. Here, we examine the developing kidney vasculature to assess its 3-dimensional structure and transcriptional heterogeneity. First, we observe that endothelial cells (ECs) grow coordinately with the kidney bud as early as E10.5, and begin to show signs of specification by E13.5 when the first arteries can be identified. We then focus on how ECs pattern and remodel with respect to the developing nephron and collecting duct epithelia. ECs circumscribe nephron progenitor populations at the distal tips of the ureteric bud (UB) tree and form stereotyped cruciform structures around each tip. Beginning at the renal vesicle (RV) stage, ECs form a continuous plexus around developing nephrons. The endothelial plexus envelops and elaborates with the maturing nephron, becoming preferentially enriched along the early distal tubule. Lastly, we perform transcriptional and immunofluorescent screens to characterize spatiotemporal heterogeneity in the kidney vasculature and identify novel regionally enriched genes. A better understanding of development of the kidney vasculature will help instruct engineering of properly vascularized ex vivo kidneys and evaluate diseased kidneys.


Asunto(s)
Embrión de Mamíferos/embriología , Células Endoteliales/metabolismo , Túbulos Renales Distales/embriología , Organogénesis/fisiología , Arteria Renal/embriología , Venas Renales , Animales , Embrión de Mamíferos/citología , Células Endoteliales/citología , Células Madre Fetales/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Túbulos Renales Distales/citología , Ratones , Arteria Renal/citología , Venas Renales/crecimiento & desarrollo , Venas Renales/metabolismo , Transcripción Genética/fisiología , Uretra/citología , Uretra/embriología
12.
Physiol Genomics ; 50(5): 343-354, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29521601

RESUMEN

The renal aldosterone-sensitive distal tubule (ASDT) is crucial for sodium reabsorption and blood pressure regulation. The ASDT consists of the late distal convoluted tubule (DCT2), connecting tubule (CNT), and collecting duct. Due to difficulties in isolating epithelial cells from the ASDT in large quantities, few transcriptome studies have been performed on this segment. Moreover, no studies exist on isolated DCT2 and CNT cells (excluding intercalated cells), and the role of aldosterone for regulating the transcriptome of these specific cell types is largely unknown. A mouse model expressing eGFP in DCT2/CNT/initial cortical collecting duct (iCCD) principal cells was exploited to facilitate the isolation of these cells in high number and purity. Combined with deep RNA sequencing technology, a comprehensive catalog of chronic aldosterone-regulated transcripts from enriched DCT2/CNT/iCCD principal cells was generated. There were 257 significantly downregulated and 290 upregulated transcripts in response to aldosterone ( P < 0.05). The RNA sequencing confirmed aldosterone regulation of well-described aldosterone targets including Sgk1 and Tsc22d3. Changes in selected transcripts such as S100a1 and Cldn4 were confirmed by RT-qPCR. The RNA sequencing showed downregulation of Nr3c2 encoding the mineralocorticoid receptor (MR), and cell line experiments showed a parallel decrease in MR protein. Furthermore, a large number of transcripts encoding transcription factors were downregulated. An extensive mRNA transcriptome reconstruction of an enriched CNT/iCCD principal cell population was also generated. The results provided a comprehensive database of aldosterone-regulated transcripts in the ASDT, allowing development of novel hypotheses for the action of aldosterone.


Asunto(s)
Aldosterona/farmacología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Análisis de Secuencia de ARN/métodos , Transcriptoma/efectos de los fármacos , Aldosterona/administración & dosificación , Aldosterona/sangre , Animales , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Túbulos Renales Colectores/citología , Túbulos Renales Distales/citología , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Urolithiasis ; 46(5): 409-418, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29383416

RESUMEN

The calcium sensing receptor (CaSR) in the distal nephron decreases the propensity for calcium stones. Here we investigate if the apical CaSR in the proximal tubule also prevents stone formation acting via regulation of apical dicarboxylate and citrate transport. Urinary citrate, partially reabsorbed as a dicarboxylate in the proximal tubule lumen, inhibits stone formation by complexing calcium. We previously demonstrated a novel apical calcium-sensitive dicarboxylate transport system in OK proximal tubule cells. This calcium-sensitive process has the potential to modulate the amount of citrate available to complex increased urinary calcium. Using isotope labeled succinate uptake in OK cells along with various pharmacologic tools we examined whether the CaSR alters apical dicarboxylate transport and through which signal transduction pathways this occurs. Our results indicate that in the proximal tubule CaSR adjusts apical dicarboxylate transport, and does so via a CaSR â†’ Gq â†’ PKC signaling pathway. Thus, the CaSR may decrease the propensity for stone formation via actions in both proximal and distal nephron segments.


Asunto(s)
Calcio/metabolismo , Ácido Cítrico/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Proximales/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Ácidos Dicarboxílicos/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Proximales/citología , Zarigüeyas , Eliminación Renal
14.
J Am Soc Nephrol ; 29(1): 57-68, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021385

RESUMEN

The renin-angiotensin-aldosterone system has an important role in the control of fluid homeostasis and BP during volume depletion. Dietary salt restriction elevates circulating angiotensin II (AngII) and aldosterone levels, increasing levels of the Cl-/HCO3- exchanger pendrin in ß-intercalated cells and the Na+-Cl- cotransporter (NCC) in distal convoluted tubules. However, the independent roles of AngII and aldosterone in regulating these levels remain unclear. In C57BL/6J mice receiving a low-salt diet or AngII infusion, we evaluated the membrane protein abundance of pendrin and NCC; assessed the phosphorylation of the mineralocorticoid receptor, which selectively inhibits aldosterone binding in intercalated cells; and measured BP by radiotelemetry in pendrin-knockout and wild-type mice. A low-salt diet or AngII infusion upregulated NCC and pendrin levels, decreased the phosphorylation of mineralocorticoid receptor in ß-intercalated cells, and increased plasma aldosterone levels. Notably, a low-salt diet did not alter BP in wild-type mice, but significantly decreased BP in pendrin-knockout mice. To dissect the roles of AngII and aldosterone, we performed adrenalectomies in mice to remove aldosterone from the circulation. In adrenalectomized mice, AngII infusion again upregulated NCC expression, but did not affect pendrin expression despite the decreased phosphorylation of mineralocorticoid receptor. By contrast, AngII and aldosterone coadministration markedly elevated pendrin levels in adrenalectomized mice. Our results indicate that aldosterone is necessary for AngII-induced pendrin upregulation, and suggest that pendrin contributes to the maintenance of normal BP in cooperation with NCC during activation of the renin-angiotensin-aldosterone system by dietary salt restriction.


Asunto(s)
Aldosterona/sangre , Angiotensina II/farmacología , Simportadores del Cloruro de Sodio/metabolismo , Transportadores de Sulfato/metabolismo , Vasoconstrictores/farmacología , Adrenalectomía , Aldosterona/farmacología , Animales , Presión Sanguínea/genética , Túbulos Renales Distales/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Receptores de Mineralocorticoides/metabolismo , Cloruro de Sodio Dietético/administración & dosificación , Transportadores de Sulfato/genética , Regulación hacia Arriba/efectos de los fármacos
15.
J Am Soc Nephrol ; 28(6): 1814-1825, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28052988

RESUMEN

The mammalian distal convoluted tubule (DCT) makes an important contribution to potassium homeostasis by modulating NaCl transport. The thiazide-sensitive Na+/Cl- cotransporter (NCC) is activated by low potassium intake and by hypokalemia. Coupled with suppression of aldosterone secretion, activation of NCC helps to retain potassium by increasing electroneutral NaCl reabsorption, therefore reducing Na+/K+ exchange. Yet the mechanisms by which DCT cells sense plasma potassium concentration and transmit the information to the apical membrane are not clear. Here, we tested the hypothesis that the potassium channel Kir4.1 is the potassium sensor of DCT cells. We generated mice in which Kir4.1 could be deleted in the kidney after the mice are fully developed. Deletion of Kir4.1 in these mice led to moderate salt wasting, low BP, and profound potassium wasting. Basolateral membranes of DCT cells were depolarized, nearly devoid of conductive potassium transport, and unresponsive to plasma potassium concentration. Although renal WNK4 abundance increased after Kir4.1 deletion, NCC abundance and function decreased, suggesting that membrane depolarization uncouples WNK kinases from NCC. Together, these results indicate that Kir4.1 mediates potassium sensing by DCT cells and couples this signal to apical transport processes.


Asunto(s)
Túbulos Renales Distales/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Potasio , Animales , Túbulos Renales Distales/citología , Ratones
16.
J Physiol Biochem ; 73(2): 199-205, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27909897

RESUMEN

Increased renal reabsorption of sodium is a significant risk factor in hypertension. An established clinical marker for essential hypertension is elevated sodium lithium countertransport (SLC) activity. NHA2 is a newly identified Na+(Li+)/H+ antiporter with potential genetic links to hypertension, which has been shown to mediate SLC activity and H+-coupled Na+(Li+) efflux in kidney-derived MDCK cells. To evaluate a putative role in sodium homeostasis, we determined the effect of dietary salt on NHA2. In murine kidney sections, NHA2 localized apically to distal convoluted (both DCT1 and 2) and connecting tubules, partially overlapping in distribution with V-ATPase, AQP2, and NCC1 transporters. Mice fed a diet high in sodium chloride showed elevated transcripts and expression of NHA2 protein. We propose a model in which NHA2 plays a dual role in salt reabsorption or secretion, depending on the coupling ion (sodium or protons). The identified novel regulation of Na+/H+ antiporter in the kidney suggests new roles in salt homeostasis and disease.


Asunto(s)
Antiportadores/metabolismo , Regulación de la Expresión Génica , Nefronas/metabolismo , Sodio en la Dieta/administración & dosificación , Animales , Antiportadores/genética , Acuaporina 2/metabolismo , Biomarcadores/metabolismo , Polaridad Celular , Hipertensión/sangre , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/patología , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Nefronas/citología , Nefronas/patología , Especificidad de Órganos , Transporte de Proteínas , ARN Mensajero/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Sodio en la Dieta/efectos adversos , ATPasas de Translocación de Protón Vacuolares/metabolismo
17.
J Biol Chem ; 291(47): 24787-24799, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27733684

RESUMEN

Hypotonic stress decreased claudin-1 and -2 expression levels in renal tubular epithelial HK-2 and Madin-Darby canine kidney cells. Here, we examined the regulatory mechanism involved in this decrease. The hypotonicity-induced decrease in claudin expression was inhibited by the following: SB202190, a p38 MAPK inhibitor, but not by U0126, a MEK inhibitor; Go6983, a protein kinase C inhibitor; or SP600125, a Jun N-terminal protein kinase inhibitor. Hypotonic stress increased transepithelial electrical resistance, which was inhibited by SB202190. The mRNA expression level of claudin-1 was decreased by hypotonic stress but that of claudin-2 was not. Hypotonic stress decreased the protein stability of claudin-1 and -2. The hypotonicity-induced decrease in claudin expression was inhibited by the following: chloroquine, a lysosome inhibitor; dynasore and monodansylcadaverine, clathrin-dependent endocytosis inhibitors; and siRNA against clathrin heavy chain. Claudin-1 and -2 were mainly distributed in the cytosol and tight junctions (TJs) in the chloroquine- and monodansylcadaverine-treated cells, respectively. Hypotonic stress decreased the phosphorylation levels of claudin-1 and -2, which were inhibited by the protein phosphatase inhibitors okadaic acid and cantharidin. Dephosphorylated mutants of claudin-1 and -2 were mainly distributed in the cytosol, which disappeared in response to hypotonic stress. In contrast, mimicking phosphorylation mutants were distributed in the TJs, which were not decreased by hypotonic stress. We suggest that hypotonic stress induces dephosphorylation, clathrin-dependent endocytosis, and degradation of claudin-1 and -2 in lysosomes, resulting in disruption of the TJ barrier in renal tubular epithelial cells.


Asunto(s)
Clatrina/metabolismo , Claudina-1/metabolismo , Claudinas/metabolismo , Regulación hacia Abajo , Endocitosis , Células Epiteliales/metabolismo , Túbulos Renales Distales/metabolismo , Presión Osmótica , Animales , Clatrina/genética , Claudina-1/genética , Claudinas/genética , Citosol/metabolismo , Perros , Humanos , Túbulos Renales Distales/citología , Células de Riñón Canino Madin Darby , Fosforilación , Uniones Estrechas/genética , Uniones Estrechas/metabolismo
18.
Sci Rep ; 6: 27192, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27273361

RESUMEN

Pharmacological blockade of the N- and L-type calcium channel lessens renal injury in kidney disease patients. The significance of specific blockade of α1 subunit of N-type calcium channel, Cav2.2, in diabetic nephropathy, however, remains to be clarified. To examine functional roles, we mated Cav2.2(-/-) mice with db/db (diabetic) mice on the C57BLKS background. Cav2.2 was localized in glomeruli including podocytes and in distal tubular cells. Diabetic Cav2.2(-/-) mice significantly reduced urinary albumin excretion, glomerular hyperfiltration, blood glucose levels, histological deterioration and systolic blood pressure (SBP) with decreased urinary catecholamine compared to diabetic Cav2.2(+/+) mice. Interestingly, diabetic heterozygous Cav2.2(+/-) mice also decreased albuminuria, although they exhibited comparable systolic blood pressure, sympathetic nerve activity and creatinine clearance to diabetic Cav2.2(+/+) mice. Consistently, diabetic mice with cilnidipine, an N-/L-type calcium channel blocker, showed a reduction in albuminuria and improvement of glomerular changes compared to diabetic mice with nitrendipine. In cultured podocytes, depolarization-dependent calcium responses were decreased by ω-conotoxin, a Cav2.2-specific inhibitor. Furthermore, reduction of nephrin by transforming growth factor-ß (TGF-ß) in podocytes was abolished with ω-conotoxin, cilnidipine or mitogen-activated protein kinase kinase inhibitor. In conclusion, Cav2.2 inhibition exerts renoprotective effects against the progression of diabetic nephropathy, partly by protecting podocytes.


Asunto(s)
Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Canales de Calcio Tipo N/genética , Nefropatías Diabéticas/tratamiento farmacológico , omega-Conotoxinas/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Ratones , Podocitos/citología , Podocitos/metabolismo , omega-Conotoxinas/farmacología
19.
Am J Physiol Renal Physiol ; 310(9): F872-84, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26823281

RESUMEN

Rats that have recovered from severe proximal tubule (PT) injury induced by uranyl acetate (UA), a toxic stimulus, developed resistance to subsequent UA treatment. We investigated cell cycle status and progression in PT cells in relation to this acquired resistance. Fourteen days after pretreatment with saline (vehicle group) or UA [acute kidney injury (AKI) group], rats were injected with UA or lead acetate (a proliferative stimulus). Cell cycle status (G0/G1/S/G2/M) was analyzed by flow cytometry. The expression of cell cycle markers, cyclin-dependent kinase inhibitors, and phenotypic markers were examined by immunohistochemistry. Cell cycle status in PT cells in the AKI group was comparable to those of the vehicle group. However, more early G1-phase cells (cyclin D1- or Ki67-) and p21+ or p27+ cells were found in the PT of the AKI group than in that of the vehicle group. UA induced G1 arrest and inhibited S phase progression with earlier dedifferentiation and less apoptosis in PT cells of the AKI group. Lead acetate induced proliferation without dedifferentiation but with delayed G0-G1 transition and inhibited S phase progression in PT cells in the AKI group. Sustained p21 and increased p27 expression in PT cells were found in the AKI group in response to UA and lead acetate. PT cells in the AKI group inhibited cell cycle progression by enhanced G1 arrest, probably via p21/p27 modulation as an injury or proliferation response, resulting in cytoresistance to rechallenge injury.


Asunto(s)
Lesión Renal Aguda/patología , Puntos de Control del Ciclo Celular , Túbulos Renales Proximales/patología , Lesión Renal Aguda/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Desdiferenciación Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/biosíntesis , Túbulos Renales Distales/citología , Túbulos Renales Distales/efectos de los fármacos , Masculino , Compuestos Organometálicos , Ratas , Ratas Sprague-Dawley , Recurrencia , Quinasas p21 Activadas/biosíntesis
20.
Kidney Int ; 89(1): 127-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26422504

RESUMEN

Dietary potassium deficiency activates thiazide-sensitive sodium chloride cotransport along the distal nephron. This may explain, in part, the hypertension and cardiovascular mortality observed in individuals who consume a low-potassium diet. Recent data suggest that plasma potassium affects the distal nephron directly by influencing intracellular chloride, an inhibitor of the with-no-lysine kinase (WNK)-Ste20p-related proline- and alanine-rich kinase (SPAK) pathway. As previous studies used extreme dietary manipulations, we sought to determine whether the relationship between potassium and NaCl cotransporter (NCC) is physiologically relevant and clarify the mechanisms involved. We report that modest changes in both dietary and plasma potassium affect NCC in vivo. Kinase assay studies showed that chloride inhibits WNK4 kinase activity at lower concentrations than it inhibits activity of WNK1 or WNK3. Also, chloride inhibited WNK4 within the range of distal cell chloride concentration. Mutation of a previously identified WNK chloride-binding motif converted WNK4 effects on SPAK from inhibitory to stimulatory in mammalian cells. Disruption of this motif in WNKs 1, 3, and 4 had different effects on NCC, consistent with the three WNKs having different chloride sensitivities. Thus, potassium effects on NCC are graded within the physiological range, which explains how unique chloride-sensing properties of WNK4 enable it to mediate effects of potassium on NCC in vivo.


Asunto(s)
Cloruros/metabolismo , Homeostasis , Potasio/sangre , Proteínas Serina-Treonina Quinasas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Distales/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Mutación , Nefronas/fisiología , Fosforilación/genética , Potasio/metabolismo , Potasio en la Dieta/administración & dosificación , Proteínas Serina-Treonina Quinasas/genética , Proteína Quinasa Deficiente en Lisina WNK 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...