Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 130(10): 1550-1564, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35430873

RESUMEN

BACKGROUND: Renal T cells contribute importantly to hypertension, but the underlying mechanism is incompletely understood. We reported that CD8Ts directly stimulate distal convoluted tubule cells (DCTs) to increase NCC (sodium chloride co-transporter) expression and salt reabsorption. However, the mechanistic basis of this pathogenic pathway that promotes hypertension remains to be elucidated. METHODS: We used mouse models of DOCA+salt (DOCA) treatment and adoptive transfer of CD8+ T cells (CD8T) from hypertensive animals to normotensive animals in in vivo studies. Co-culture of mouse DCTs and CD8Ts was used as in vitro model to test the effect of CD8T activation in promoting NCC-mediated sodium retention and to identify critical molecular players contributing to the CD8T-DCT interaction. Interferon (IFNγ)-KO mice and mice receiving renal tubule-specific knockdown of PDL1 were used to verify in vitro findings. Blood pressure was continuously monitored via radio-biotelemetry, and kidney samples were saved at experimental end points for analysis. RESULTS: We identified critical molecular players and demonstrated their roles in augmenting the CD8T-DCT interaction leading to salt-sensitive hypertension. We found that activated CD8Ts exhibit enhanced interaction with DCTs via IFN-γ-induced upregulation of MHC-I and PDL1 in DCTs, thereby stimulating higher expression of NCC in DCTs to cause excessive salt retention and progressive elevation of blood pressure. Eliminating IFN-γ or renal tubule-specific knockdown of PDL1 prevented T cell homing into the kidney, thereby attenuating hypertension in 2 different mouse models. CONCLUSIONS: Our results identified the role of activated CD8Ts in contributing to increased sodium retention in DCTS through the IFNγ-PDL1 pathway. These findings provide a new mechanism for T cell involvement in the pathogenesis of hypertension and reveal novel therapeutic targets.


Asunto(s)
Acetato de Desoxicorticosterona , Hipertensión , Animales , Linfocitos T CD8-positivos/metabolismo , Acetato de Desoxicorticosterona/metabolismo , Acetato de Desoxicorticosterona/farmacología , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Ratones , Sodio/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Cloruro de Sodio Dietético
2.
Proc Natl Acad Sci U S A ; 116(8): 3155-3160, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30718414

RESUMEN

Calcineurin is a calcium/calmodulin-regulated phosphatase known for its role in activation of T cells following engagement of the T cell receptor. Calcineurin inhibitors (CNIs) are widely used as immunosuppressive agents; common adverse effects of CNIs are hypertension and hyperkalemia. While previous studies have implicated activation of the Na-Cl cotransporter (NCC) in the renal distal convoluted tubule (DCT) in this toxicity, the molecular mechanism of this effect is unknown. The renal effects of CNIs mimic the hypertension and hyperkalemia that result from germ-line mutations in with-no-lysine (WNK) kinases and the Kelch-like 3 (KLHL3)-CUL3 ubiquitin ligase complex. WNK4 is an activator of NCC and is degraded by binding to KLHL3 followed by WNK4's ubiquitylation and proteasomal degradation. This binding is prevented by phosphorylation of KLHL3 at serine 433 (KLHL3S433-P) via protein kinase C, resulting in increased WNK4 levels and increased NCC activity. Mechanisms mediating KLHL3S433-P dephosphorylation have heretofore been unknown. We now demonstrate that calcineurin expressed in DCT is a potent KLHL3S433-P phosphatase. In mammalian cells, the calcium ionophore ionomycin, a calcineurin activator, reduces KLHL3S433-P levels, and this effect is reversed by the calcineurin inhibitor tacrolimus and by siRNA-mediated knockdown of calcineurin. In vivo, tacrolimus increases levels of KLHL3S433-P, resulting in increased levels of WNK4, phosphorylated SPAK, and NCC. Moreover, tacrolimus attenuates KLHL3-mediated WNK4 ubiquitylation and degradation, while this effect is absent in KLHL3 with S433A substitution. Additionally, increased extracellular K+ induced calcineurin-dependent dephosphorylation of KLHL3S433-P These findings demonstrate that KLHL3S433-P is a calcineurin substrate and implicate increased KLHL3 phosphorylation in tacrolimus-induced pathologies.


Asunto(s)
Proteínas Portadoras/genética , Hipertensión/genética , Proteínas Serina-Treonina Quinasas/genética , Insuficiencia Renal/genética , Proteínas Adaptadoras Transductoras de Señales , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Calcineurina/genética , Inhibidores de la Calcineurina/administración & dosificación , Proteínas Cullin/genética , Regulación de la Expresión Génica/efectos de los fármacos , Mutación de Línea Germinal/genética , Humanos , Hiperpotasemia/genética , Hiperpotasemia/metabolismo , Hiperpotasemia/patología , Hipertensión/metabolismo , Hipertensión/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Ratones , Proteínas de Microfilamentos , Complejos Multiproteicos/genética , Fosforilación , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/tratamiento farmacológico , Insuficiencia Renal/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Tacrolimus/toxicidad , Ubiquitinación
3.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768847

RESUMEN

Gitelman and Bartter syndromes are rare inherited diseases that belong to the category of renal tubulopathies. The genes associated with these pathologies encode electrolyte transport proteins located in the nephron, particularly in the Distal Convoluted Tubule and Ascending Loop of Henle. Therefore, both syndromes are characterized by alterations in the secretion and reabsorption processes that occur in these regions. Patients suffer from deficiencies in the concentration of electrolytes in the blood and urine, which leads to different systemic consequences related to these salt-wasting processes. The main clinical features of both syndromes are hypokalemia, hypochloremia, metabolic alkalosis, hyperreninemia and hyperaldosteronism. Despite having a different molecular etiology, Gitelman and Bartter syndromes share a relevant number of clinical symptoms, and they have similar therapeutic approaches. The main basis of their treatment consists of electrolytes supplements accompanied by dietary changes. Specifically for Bartter syndrome, the use of non-steroidal anti-inflammatory drugs is also strongly supported. This review aims to address the latest diagnostic challenges and therapeutic approaches, as well as relevant recent research on the biology of the proteins involved in disease. Finally, we highlight several objectives to continue advancing in the characterization of both etiologies.


Asunto(s)
Síndrome de Bartter/patología , Síndrome de Gitelman/patología , Túbulos Renales Distales/patología , Asa de la Nefrona/patología , Equilibrio Hidroelectrolítico/fisiología , Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Síndrome de Bartter/terapia , Electrólitos/análisis , Electrólitos/uso terapéutico , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Síndrome de Gitelman/terapia , Humanos , Hiperaldosteronismo/patología , Hipercalciuria/patología , Hipopotasemia/patología , Hiponatremia/patología , Nefrocalcinosis/patología , Defectos Congénitos del Transporte Tubular Renal/patología
4.
J Mol Cell Cardiol ; 138: 66-74, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31758962

RESUMEN

There is controversy regarding whether excess FGF23 causes left ventricular hypertrophy (LVH) directly through activation of fibroblast growth factor receptor 4 (FGFR4) in cardiomyocytes or indirectly through reductions in soluble Klotho (sK). We investigated the respective roles of myocardial FGFR4 and sKL in mediating FGF23-induced LVH using mouse genetic and pharmacological approaches. To investigate a direct role of myocardial FGFR4 in mediating the cardiotoxic effects of excess circulating FGF23, we administered rFGF23 to mice with cardiac-specific loss of FGFR4 (FGFR4 heart-cKO). We tested a model of sKL deficiency, hypertension and LVH created by the conditional deletion of FGFR1 in the renal distal tubule (FGFR1DT cKO mice). The cardioprotective effects of sKL in both mouse models was assessed by the systemic administration of recombinant sKL. We confirmed that FGF23 treatment activates PLCγ in the heart and induces LVH in the absence of membrane α-Klotho. Conditional deletion of FGFR4 in the myocardium prevented rFGF23-induced LVH in mice, establishing direct cardiotoxicity of FGF23 through activation of FGFR4. Recombinant sKL administration prevented LVH, but not HTN, in FGFR1DT cKO mice, consistent with direct cardioprotective effects. Co-administration of recombinant sKL with FGF23 in culture inhibited rFGF23-induced p-PLCγ signaling. Thus, FGF23 ability to include LVH represents a balance between FGF23 direct cardiac activation of FGFR4 and the modulating effects of circulating sKL to alter FGF23-dependent myocardial signaling pathways.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Citoprotección , Factor-23 de Crecimiento de Fibroblastos , Eliminación de Gen , Células HEK293 , Humanos , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Túbulos Renales Distales/patología , Proteínas Klotho , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Solubilidad
5.
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
6.
FASEB J ; 33(6): 7192-7201, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30848940

RESUMEN

Hypomagnesemia (blood Mg2+ concentration <0.7 mM) is a common electrolyte disorder in patients with type 2 diabetes (T2D), but the etiology remains largely unknown. In patients with T2D, reduced blood Mg2+ levels are associated with an increased decline in renal function, independent of glycemic control and hypertension. To study the underlying mechanism of this phenomenon, we investigated the renal effects of hypomagnesemia in high-fat-diet (HFD)-fed mice. In mice fed a low dietary Mg2+, the HFD resulted in severe hypomagnesemia within 4 wk. Renal or intestinal Mg2+ wasting was not observed after 16 wk on the diets. Despite the absence of urinary or fecal Mg2+ loss, the HFD induced a reduction in the mRNA expression transient receptor potential melastatin type 6 in both the kidney and colon. mRNA expression of distal convoluted tubule (DCT)-specific genes was down-regulated by the LowMg-HFD, indicating atrophy of the DCT. The low dietary Mg2+ resulted in severe HFD-induced proximal tubule phospholipidosis, which was absent in mice on a NormalMg-HFD. This was accompanied by albuminuria, moderate renal damage, and alterations in renal energy metabolism, including enhanced gluconeogenesis and cholesterol synthesis. In conclusion, this study shows that hypomagnesemia is a consequence of diet-induced obesity and insulin resistance. Moreover, hypomagnesemia induces major structural changes in the diabetic kidney, including proximal tubular phospholipidosis, providing a novel mechanism for the increased renal decline in patients with hypomagnesemic T2D.-Kurstjens, S., Smeets, B., Overmars-Bos, C., Dijkman, H. B., den Braanker, D. J. W., de Bel, T., Bindels, R. J. M., Tack, C. J. J., Hoenderop, J. G. J., de Baaij, J. H. F. Renal phospholipidosis and impaired magnesium handling in high-fat-diet-fed mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Túbulos Renales Distales/metabolismo , Túbulos Renales Proximales/metabolismo , Deficiencia de Magnesio/metabolismo , Magnesio/metabolismo , Obesidad/metabolismo , Fosfolípidos/metabolismo , Albuminuria/etiología , Animales , Atrofia , Líquidos Corporales/química , Metabolismo Energético , Heces/química , Resistencia a la Insulina , Túbulos Renales Distales/patología , Túbulos Renales Proximales/patología , Magnesio/administración & dosificación , Magnesio/farmacocinética , Deficiencia de Magnesio/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Obesidad/complicaciones , ARN Mensajero/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales Catiónicos TRPM/biosíntesis , Canales Catiónicos TRPM/genética
7.
Nephrol Dial Transplant ; 35(3): 411-432, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31436795

RESUMEN

BACKGROUND: Loss-of-function mutations in the sodium chloride (NaCl) co-transporter (NCC) of the renal distal convoluted tubule (DCT) cause Gitelman syndrome with hypokalemic alkalosis, hypomagnesemia and hypocalciuria. Since Gitelman patients are usually diagnosed around adolescence, we tested the idea that a progressive regression of the DCT explains the late clinical onset of the syndrome. METHODS: NCC wild-type and knockout (ko) mice were studied at Days 1, 4 and 10 and 6 weeks after birth using blood plasma analysis and morphological and biochemical methods. RESULTS: Plasma aldosterone levels and renal renin messenger RNA expression were elevated in NCC ko mice during the first days of life. In contrast, plasma ion levels did not differ between genotypes at age 10 days, but a significant hypomagnesemia was observed in NCC ko mice at 6 weeks. Immunofluorescent detection of parvalbumin (an early DCT marker) revealed that the fractional cortical volume of the early DCT is similar for mice of both genotypes at Day 4, but is significantly lower at Day 10 and is almost zero at 6 weeks in NCC ko mice. The DCT atrophy correlates with a marked reduction in the abundance of the DCT-specific Mg2+ channel TRPM6 (transient receptor potential cation channel subfamily M member 6) and an increased proteolytic activation of the epithelial Na+ channel (ENaC). CONCLUSION: After an initial outgrowth, DCT development lags behind in NCC ko mice. The impaired DCT development associates at Day 1 and Day 10 with elevated renal renin and plasma aldosterone levels and activation of ENaC, respectively, suggesting that Gitelman syndrome might be present much earlier in life than is usually expected. Despite an early downregulation of TRPM6, hypomagnesemia is a rather late symptom.


Asunto(s)
Síndrome de Bartter/patología , Síndrome de Gitelman/patología , Hiperaldosteronismo/patología , Túbulos Renales Distales/patología , Magnesio/metabolismo , Simportadores del Cloruro de Sodio/fisiología , Sodio/metabolismo , Animales , Síndrome de Bartter/etiología , Síndrome de Bartter/metabolismo , Síndrome de Gitelman/etiología , Síndrome de Gitelman/metabolismo , Hiperaldosteronismo/etiología , Hiperaldosteronismo/metabolismo , Túbulos Renales Distales/metabolismo , Ratones , Ratones Noqueados , Renina/metabolismo , Canales Catiónicos TRPM/metabolismo
8.
Pediatr Nephrol ; 35(10): 1815-1824, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31664557

RESUMEN

Bartter and Gitelman syndromes are rare inherited tubulopathies characterized by hypokalaemic, hypochloraemic metabolic alkalosis. They are caused by mutations in at least 7 genes involved in the reabsorption of sodium in the thick ascending limb (TAL) of the loop of Henle and/or the distal convoluted tubule (DCT). Different subtypes can be distinguished and various classifications have been proposed based on clinical symptoms and/or the underlying genetic cause. Yet, the clinical phenotype can show remarkable variability, leading to potential divergences between classifications. These problems mostly relate to uncertainties over the role of the basolateral chloride exit channel CLCNKB, expressed in both TAL and DCT and to what degree the closely related paralogue CLCNKA can compensate for the loss of CLCNKB function. Here, we review what is known about the physiology of the transport proteins involved in these disorders. We also review the various proposed classifications and explain why a gene-based classification constitutes a pragmatic solution.


Asunto(s)
Síndrome de Bartter/clasificación , Canales de Cloruro/genética , Síndrome de Gitelman/clasificación , Sodio/metabolismo , Síndrome de Bartter/genética , Síndrome de Bartter/patología , Canales de Cloruro/metabolismo , Síndrome de Gitelman/genética , Síndrome de Gitelman/patología , Humanos , Túbulos Renales Distales/patología , Asa de la Nefrona/patología , Mutación , Reabsorción Renal/genética
9.
Nephrology (Carlton) ; 25(2): 179-186, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31074544

RESUMEN

OBJECTIVES: The presence of the peritubular capillaritis and its extent are important for diagnosis of the antibody-mediated rejection in kidneys. However, it is recommended that peritubular capillaritis should only be scored in the cortex. This study aims to focus on peritubular capillaritis scoring both in the cortex and the medulla to understand the value of the medulla in the diagnosis of antibody-mediated rejection. METHODS: Fifty-one allograft renal biopsy were re-evaluated for peritubular capillaritis, C4d and acute tubular injury, separately for the cortex and the medulla according to the Banff. RESULTS: Seventeen cases (33.3%) had peritubular capillaritis both in the cortex and the medulla and three (5.9%) cases had peritubular capillaritis only in the cortex while five (9.8%) cases had only in the medulla. Eighteen (35%) of the cases had C4d staining both in the cortex and the medulla and 14 (27.5%) cases had C4d positivity only in the cortex and 18 (35.3%) cases only in the medulla. Twenty-three (45%) cases had acute tubular injury both in the cortex and the medulla and 31 (60.7%) cases had acute tubular injury only in the cortex and 23 (45.1%) cases had only in the medulla. The sensitivity, specificity, positive and negative predictive values of medullar peritubular capillaritis predicting cortical peritubular capillaritis were 85.7%, 86.7%, 81.8% and 89.7%, respectively. CONCLUSION: In case of absence of the cortical tissue, medulla can be used as a reference for antibody-mediated rejection considering the morphological features, results of donor-specific antibody and renal function tests.


Asunto(s)
Rechazo de Injerto , Corteza Renal , Trasplante de Riñón , Túbulos Renales Distales , Adulto , Biopsia/métodos , Capilares/patología , Complemento C4b/análisis , Femenino , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/inmunología , Humanos , Corteza Renal/inmunología , Corteza Renal/patología , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Túbulos Renales Distales/irrigación sanguínea , Túbulos Renales Distales/inmunología , Túbulos Renales Distales/patología , Masculino , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Inmunología del Trasplante
10.
Am J Pathol ; 188(9): 2120-2138, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29935167

RESUMEN

The distal tubule (DT) helps regulate blood pressure and electrolytes. We describe a novel, autosomal recessive, morphofunctional DT abnormality in inbred mice evident as columnar alternations and age-related cystic changes. This abnormality developed in both sexes of DBA/2Cr. Similar phenotypes were observed in A/J, C3H/He, DBA/1J, and FVB/N strains, but not in AKR/N, BALB/c, or C57BL/6N strains. In DBA/2Cr, abnormal DT localized to straight and convoluted segments and showed IL-36α DT injury marker expression. However, DT epithelial proliferation, examined by bromodeoxyuridine incorporation, was not remarkably altered with the progression of abnormality. Abnormal DT epithelial cells in DBA/2Cr displayed elongated primary cilia, loose intercellular adhesions, and numerous vesicles with altered localization of CD9, Na+/K+ATPase, and E-cadherin, indicating altered cell function, adhesion, and polarity. DBA/2Cr-type D12Mit182-D12Mit83 was identified as a candidate locus designated DBA/2 renal cyst (drecy). Within drecy, the gene regulated by estrogen in breast cancer protein (Greb1) transcript variant 2 was significantly up-regulated in DBA/2Cr kidney versus C57BL/6N. Greb1 localized to DT cytoplasm in C57BL/6 and to cytoplasm and nucleus in DBA/2Cr. Greb1-overexpressing M-1 kidney cells showed an altered epithelial-mesenchyme phenotype. B6.D2-(D12Mit182-D12Mit83) congenic mice carrying drecy did not show DT abnormalities, whereas DBA/2Cr × B6.D2-(D12Mit182-D12Mit83) mice did. Identification of this novel DT abnormality regulated by a DBA/2Cr mouse chromosome 12-derived locus and additional genetic factors improve the understanding of DT pathogenesis.


Asunto(s)
Cromosomas de los Mamíferos , Susceptibilidad a Enfermedades , Marcadores Genéticos , Enfermedades Renales/patología , Túbulos Renales Distales/patología , Animales , Femenino , Perfilación de la Expresión Génica , Enfermedades Renales/genética , Túbulos Renales Distales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
11.
Cell Tissue Res ; 378(2): 301-317, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31256287

RESUMEN

The aim was to explore the influence of experimental diabetes mellitus type 1 (DM1) and potential protective/deleterious effects of different dietary n-6/n-3 PUFA ratios on renal phospholipid composition and pathological changes caused by DM1. Male Wistar rats were injected with 55 mg/kg streptozotocin or citrate buffer (control group). Control (C) and diabetic groups (STZ) were fed with n-6/n-3 ratio of ≈ 7, STZ + N6 with n-6/n-3 ratio ≈ 60 and STZ + DHA with n-6/n-3 ratio of ≈ 1 containing 16% EPA and 19% DHA. Tissues were harvested 30 days after DM1 induction. Blood and kidneys were collected and analysed for phospholipid fatty acid composition, pathohystological changes, ectopic lipid accumulation and expression of VEGF, NF-kB and special AT-rich sequence-binding protein-1 (SATB1). Pathological changes were studied also by using transmission electron microscopy, after immunostaining for VEGF. Substantial changes in renal phospholipid fatty acid composition resulted from DM1 and dietary PUFA manipulation. Extensive vacuolization of distal tubular cells (DTCs) was found in DM1, but it was attenuated in the STZ + DHA group, in which the highest renal NF-kB expression was observed. The ectopic lipid accumulation was observed in proximal tubular cells (PTCs) of all diabetic animals, but it was worsened in the STZ + N6 group. In DM1, we found disturbance of VEGF-transporting vesicular PTCs system, which was substantially worsened in STZ + DHA and STZ + N6. Results have shown that the early phase of DN is characterized with extent damage and vacuolization of DTCs, which could be attenuated by DHA/EPA supplementation. We concluded that dietary fatty acid composition can strongly influence the outcomes of DN.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Túbulos Renales Distales , Animales , Diabetes Mellitus Experimental , Suplementos Dietéticos , Ácidos Grasos/metabolismo , Proteínas de Homeodominio/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Masculino , Fosfolípidos/metabolismo , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
J Am Soc Nephrol ; 29(2): 416-422, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29114040

RESUMEN

Podocytes contain an intricate actin cytoskeleton that is essential for the specialized function of this cell type in renal filtration. Serum response factor (SRF) is a master transcription factor for the actin cytoskeleton, but the in vivo expression and function of SRF in podocytes are unknown. We found that SRF protein colocalizes with podocyte markers in human and mouse kidneys. Compared with littermate controls, mice in which the Srf gene was conditionally inactivated with NPHS2-Cre exhibited early postnatal proteinuria, hypoalbuminemia, and azotemia. Histologic changes in the mutant mice included glomerular capillary dilation and mild glomerulosclerosis, with reduced expression of multiple canonical podocyte markers. We also noted tubular dilation, cell proliferation, and protein casts as well as reactive changes in mesangial cells and interstitial inflammation. Ultrastructure analysis disclosed foot process effacement with loss of slit diaphragms. To ascertain the importance of SRF cofactors in podocyte function, we disabled the myocardin-related transcription factor A and B genes. Although loss of either SRF cofactor alone had no observable effect in the kidney, deficiency of both recapitulated the Srf-null phenotype. These results establish a vital role for SRF and two SRF cofactors in the maintenance of podocyte structure and function.


Asunto(s)
Actinas/metabolismo , Podocitos/metabolismo , Podocitos/ultraestructura , Factor de Respuesta Sérica/fisiología , Transactivadores/genética , Factores de Transcripción/genética , Actinina/genética , Actinas/genética , Animales , Citoesqueleto , Dilatación Patológica/genética , Femenino , Humanos , Túbulos Renales Distales/patología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Noqueados , Podocitos/fisiología , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Represoras/genética , Factor de Respuesta Sérica/genética , Proteínas WT1
13.
Kidney Int ; 93(3): 580-588, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29129401

RESUMEN

The tight junction proteins claudin-10 and -16 are crucial for the paracellular reabsorption of cations along the thick ascending limb of Henle's loop in the kidney. In patients, mutations in CLDN16 cause familial hypomagnesemia with hypercalciuria and nephrocalcinosis, while mutations in CLDN10 impair kidney function. Mice lacking claudin-16 display magnesium and calcium wasting, whereas absence of claudin-10 results in hypermagnesemia and interstitial nephrocalcinosis. In order to study the functional interdependence of claudin-10 and -16 we generated double-deficient mice. These mice had normal serum magnesium and urinary excretion of magnesium and calcium and showed polyuria and sodium retention at the expense of increased renal potassium excretion, but no nephrocalcinosis. Isolated thick ascending limb tubules of double mutants displayed a complete loss of paracellular cation selectivity and functionality. Mice lacking both claudin-10 and -16 in the thick ascending limb recruited downstream compensatory mechanisms and showed hypertrophic distal convoluted tubules with changes in gene expression and phosphorylation of ion transporters in this segment, presumably triggered by the mild decrease in serum potassium. Thus, severe individual phenotypes in claudin-10 and claudin-16 knockout mice are corrected by the additional deletion of the other claudin.


Asunto(s)
Claudinas/deficiencia , Hipercalciuria/prevención & control , Túbulos Renales Distales/metabolismo , Asa de la Nefrona/metabolismo , Deficiencia de Magnesio/prevención & control , Animales , Calcio/metabolismo , Claudinas/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Predisposición Genética a la Enfermedad , Hipercalciuria/genética , Hipercalciuria/metabolismo , Hipercalciuria/fisiopatología , Túbulos Renales Distales/patología , Túbulos Renales Distales/fisiopatología , Asa de la Nefrona/patología , Asa de la Nefrona/fisiopatología , Magnesio/metabolismo , Deficiencia de Magnesio/genética , Deficiencia de Magnesio/metabolismo , Deficiencia de Magnesio/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrocalcinosis/genética , Nefrocalcinosis/metabolismo , Nefrocalcinosis/fisiopatología , Nefrocalcinosis/prevención & control , Fenotipo , Sodio/metabolismo
14.
Kidney Blood Press Res ; 43(2): 513-521, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29627839

RESUMEN

BACKGROUND/AIMS: Primary distal renal tubular acidosis (dRTA) in children is a rare genetic disorder, and three causative mutated genes have been identified: SLC4A1, ATP6V1B1, and ATP6V0A4. We analyzed the prevalence and phenotypic differences of genetic mutations in children with dRTA. METHODS: A total of 17 children with dRTA were enrolled in the study. All patients underwent genetic testing for all three candidate genes. RESULTS: Pathogenic mutations, including six novel mutations, were detected in 15 (88.2%) patients: dominant SLC4A1 mutations in ten (58.8%) patients, recessive ATP6V0A4 mutations in three (17.6%) patients, and recessive ATP6V1B1 mutations in two (11.8%) patients. Compared to other patients, patients with SLC4A1 mutations showed an older age of onset (3.7 ± 2.6 years) and less severe metabolic acidosis at initial presentation. All patients developed nephrocalcinosis, and sensorineural hearing loss was observed in two patients with ATP6V1B1 mutations. Three (17.6%) patients had decreased renal function (chronic kidney disease stage 2), and five (29.4%) patients had persistent growth retardation at the last follow-up. Long-term prognosis showed no genotype-phenotype correlation. CONCLUSIONS: SLC4A1 is the most common defective gene in Korean children with dRTA. Patients with SLC4A1 mutations show later onset and milder disease severity. Long-term follow-up of hearing ability, renal function, and growth is necessary for patients with dRTA.


Asunto(s)
Acidosis Tubular Renal/genética , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Estudios de Asociación Genética , Acidosis Tubular Renal/patología , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Crecimiento/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Túbulos Renales Distales/patología , Masculino , Mutación , Nefrocalcinosis/genética , República de Corea , ATPasas de Translocación de Protón Vacuolares/genética
15.
J Am Soc Nephrol ; 28(9): 2597-2606, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28442491

RESUMEN

Aberrant activation of with no lysine (WNK) kinases causes familial hyperkalemic hypertension (FHHt). Thiazide diuretics treat the disease, fostering the view that hyperactivation of the thiazide-sensitive sodium-chloride cotransporter (NCC) in the distal convoluted tubule (DCT) is solely responsible. However, aberrant signaling in the aldosterone-sensitive distal nephron (ASDN) and inhibition of the potassium-excretory renal outer medullary potassium (ROMK) channel have also been implicated. To test these ideas, we introduced kinase-activating mutations after Lox-P sites in the mouse Stk39 gene, which encodes the terminal kinase in the WNK signaling pathway, Ste20-related proline-alanine-rich kinase (SPAK). Renal expression of the constitutively active (CA)-SPAK mutant was specifically targeted to the early DCT using a DCT-driven Cre recombinase. CA-SPAK mice displayed thiazide-treatable hypertension and hyperkalemia, concurrent with NCC hyperphosphorylation. However, thiazide-mediated inhibition of NCC and consequent restoration of sodium excretion did not immediately restore urinary potassium excretion in CA-SPAK mice. Notably, CA-SPAK mice exhibited ASDN remodeling, involving a reduction in connecting tubule mass and attenuation of epithelial sodium channel (ENaC) and ROMK expression and apical localization. Blocking hyperactive NCC in the DCT gradually restored ASDN structure and ENaC and ROMK expression, concurrent with the restoration of urinary potassium excretion. These findings verify that NCC hyperactivity underlies FHHt but also reveal that NCC-dependent changes in the driving force for potassium secretion are not sufficient to explain hyperkalemia. Instead, a DCT-ASDN coupling process controls potassium balance in health and becomes aberrantly activated in FHHt.


Asunto(s)
Hidroclorotiazida/farmacología , Túbulos Renales Distales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Aldosterona/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Hidroclorotiazida/uso terapéutico , Túbulos Renales Distales/metabolismo , Ratones , Natriuresis/efectos de los fármacos , Fosforilación , Potasio/orina , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/tratamiento farmacológico , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/orina , Transducción de Señal , Inhibidores de los Simportadores del Cloruro de Sodio/uso terapéutico
16.
J Hum Genet ; 62(2): 335-337, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27784896

RESUMEN

Gitelman syndrome (GS) is an autosomal recessive renal tubulopathy characterized by hypokalemic metabolic alkalosis with hypocalciuria and hypomagnesemia. GS clinical symptoms range from mild weakness to muscular cramps, paralysis or even sudden death as a result of cardiac arrhythmia. GS is caused by loss-of-function mutations in the solute carrier family 12 member 3 (SLC12A3) gene, but molecular mechanisms underlying such a wide range of symptoms are poorly understood. Here we report cryptic exon activation in SLC12A3 intron 12 in a clinically asymptomatic GS, resulting from an intronic mutation c.1669+297 T>G that created a new acceptor splice site. The cryptic exon was sandwiched between the L3 transposon upstream and a mammalian interspersed repeat downstream, possibly contributing to inclusion of the cryptic exon in mature transcripts. The mutation was identified by targeted next-generation sequencing of candidate genes in GS patients with missing pathogenic SLC12A3 alleles. Taken together, this work illustrates the power of next-generation sequencing to identify causal mutations in intronic regions in asymptomatic individuals at risk of developing potentially fatal disease complications, improving clinical management of these cases.


Asunto(s)
Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Túbulos Renales Distales/patología , Secuencia de Bases , Preescolar , Exones/genética , Femenino , Síndrome de Gitelman/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Intrones/genética , Mutación/genética , Análisis de Secuencia de ADN , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
17.
Glycoconj J ; 34(2): 267-273, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28205070

RESUMEN

Disialosyl globopentaosylceramide (DSGb5) is a ganglioside originally isolated from tissue extracts of renal cell carcinoma (RCC) with metastasis. Previous in vitro experiments have suggested that DSGb5 promotes metastasis by enhancing the migration of RCC cells and downregulating NK cell cytotoxicity against RCC cells. In this study, we investigated the clinicopathological significance of DSGb5 expression in RCC and outcomes of RCC patients. A total of 156 RCC patients who underwent surgical treatments at our hospital from January 2007 through December 2012 were analyzed in this study. The expression of DSGb5 in RCC specimens was examined by immunohistochemical staining with monoclonal antibody 5F3. The immunostaining intensity of RCC tissues was assessed in comparison with that in benign renal tubules as an internal positive control. The relationship between DSGb5 expression and clinicopathological characteristics was investigated and recurrence free survival following surgery was evaluated. Microvascular invasion was observed in 68% (n = 19/28) and in 45% (n = 58/128) of the DSGb5 high expression group and low expression group, respectively (p = 0.031). Of 156 patients with a median follow up of 51 months, 18 patients (12%) developed metastasis following surgery. Patients in the DSGb5 high expression group showed significantly lower recurrence-free survival as compared with those in the DSGb5 low expression group (log-rank P = 0.047). In the present study, DSGb5 expression was associated with microvascular invasion in RCC tissues, and patients with DSGb5 high expression showed significantly lower recurrence-free survival rates. These findings suggest that DSGb5 expressed in RCC is correlated with metastasis and is a potential predictor for identifying patients who experience metastasis after surgery.


Asunto(s)
Carcinoma de Células Renales , Regulación Neoplásica de la Expresión Génica , Globósidos/biosíntesis , Neoplasias Renales , Túbulos Renales Distales , Anciano , Anticuerpos Monoclonales de Origen Murino/química , Antineoplásicos Inmunológicos/química , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/cirugía , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Neoplasias Renales/metabolismo , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Neoplasias Renales/cirugía , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Tasa de Supervivencia
18.
Curr Opin Pediatr ; 29(2): 179-186, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27906863

RESUMEN

PURPOSE OF REVIEW: The clinical presentations of Bartter's syndrome and Gitelman's syndrome will be reviewed including two most recently described hypokalemic salt-losing tubulopathies. By taking the quite heterogeneous presentations and the apparently different pathophysiologies as the basis, the applicability of the physiologic classification has been tested. RECENT FINDINGS: According to the physiologic approach, salt-losing tubulopathies can be divided into two major groups (with completely different tubular defects): first, disorders of the thick ascending limb of Henle's loop (loop disorders); second, disorders of the distal convolute tubule (DCT disorders). A combination of these two groups with complety different tubular defects will finally lead to a third group: the combined loop/DCT disorders. On the basis of pharmacologic tests (pharmacotyping), it appears that the Bartter's syndrome V belongs to the DCT group, whereas the most recently described transient antenatal Bartter's syndrome best fits in the group with the loop and DCT combination.Besides secondary hyperaldosteronism, loop disorders present a whole spectrum of (secondary) pathophysiologic characteristics with significant diagnostic and therapeutic impact, such as polyhydramnios, hyperprostaglandinuria, nephrogenic diabetes insipidus, and nephrocalcinosis. Recent reports indicate that neonatal hyperparathyroidism has also to be added to the clinical presentation of isolated loop disorders. SUMMARY: As long as gene therapy is not available, the overall therapeutic management follows the clinical presentation, which leads to the underlying pathophysiology of renal salt wasting. Thus, when dealing with Bartter's syndrome and Gitelman's syndrome, the correct physiologic and pharmacologic characterization appears to be essential for a sound diagnostic and therapeutic patient management.


Asunto(s)
Síndrome de Bartter/diagnóstico , Síndrome de Bartter/terapia , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/terapia , Túbulos Renales Distales/patología , Síndrome de Bartter/genética , Diagnóstico Diferencial , Femenino , Predisposición Genética a la Enfermedad , Síndrome de Gitelman/genética , Humanos , Túbulos Renales Distales/metabolismo , Masculino , Medición de Riesgo , Sodio/metabolismo
19.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28800174

RESUMEN

Exposure to drugs often results in toxicity in the kidney which represents the major control system maintaining homeostasis of the body and thus is especially susceptible to xenobiotics. Nephrotoxicity is a life-threatening side-effect of nonsteroidal anti-inflammatory drugs (NSAIDs). Diclofenac is one of the most frequently prescribed NSAIDs and have been reported to cause multiple organs damage. Curcumin (CUR) exhibits nephroprotective properties. Therefore, rats were divided into four groups; rats of groups 3 and 4 received diclofenac (100 mg/kg, i.m.), whereas rats of groups 2 and 4 received CUR (100 mg/kg, p.o.) for 3 days. Diclofenac revealed a significant increase in urea and creatinine levels and malondialdehyde concentration and marked reduction in catalase activity and reduced glutathione concentration. Histopathologically, diclofenac produced fatty changes and eosinophilic casts were detected in the renal tubules, those were attenuated by administration of CUR prior diclofenac.


Asunto(s)
Curcumina/farmacología , Diclofenaco/efectos adversos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Túbulos Renales Distales/metabolismo , Animales , Diclofenaco/farmacología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Túbulos Renales Distales/patología , Masculino , Ratas
20.
J Biol Chem ; 290(2): 861-71, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25378394

RESUMEN

TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.


Asunto(s)
Inflamación/metabolismo , FN-kappa B/metabolismo , Nefritis/metabolismo , Receptores de Interleucina-17/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Inflamación/etiología , Inflamación/patología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , FN-kappa B/genética , Nefritis/etiología , Nefritis/patología , Multimerización de Proteína , Ratas , Receptores de Interleucina-17/química , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/química , Transducción de Señal/genética , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA