Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
JCI Insight ; 8(3)2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36626229

RESUMEN

Preterm birth results in low nephron endowment and increased risk of acute kidney injury (AKI) and chronic kidney disease (CKD). To understand the pathogenesis of AKI and CKD in preterm humans, we generated potentially novel mouse models with a 30%-70% reduction in nephron number by inhibiting or deleting Ret tyrosine kinase in the developing ureteric bud. These mice developed glomerular and tubular hypertrophy, followed by the transition to CKD, recapitulating the renal pathological changes seen in humans born preterm. We injected neonatal mice with gentamicin, a ubiquitous nephrotoxic exposure in preterm infants, and detected more severe proximal tubular injury in mice with low nephron number compared with controls with normal nephron number. Mice with low nephron number had reduced proliferative repair with more rapid development of CKD. Furthermore, mice had more profound inflammation with highly elevated levels of MCP-1 and CXCL10, produced in part by damaged proximal tubules. Our study directly links low nephron endowment with postnatal renal hypertrophy, which in this model is maladaptive and results in CKD. Underdeveloped kidneys are more susceptible to gentamicin-induced AKI, suggesting that AKI in the setting of low nephron number is more severe and further increases the risk of CKD in this vulnerable population.


Asunto(s)
Lesión Renal Aguda , Nacimiento Prematuro , Insuficiencia Renal Crónica , Animales , Femenino , Humanos , Ratones , Lesión Renal Aguda/patología , Gentamicinas , Hipertrofia/patología , Recien Nacido Prematuro , Riñón/patología , Nefronas/patología , Nacimiento Prematuro/patología , Insuficiencia Renal Crónica/patología
2.
Kidney Int ; 99(5): 1057-1059, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33549589
3.
J Clin Invest ; 129(6): 2201-2204, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31107247

RESUMEN

Excessive excretion of oxalate in the urine results in the formation of calcium oxalate crystals and subsequent kidney stone formation. Severe forms of hyperoxaluria, including genetic forms and those that result from ethylene glycol poisoning, can result in end-stage renal disease. Therapeutic interventions are limited and often rely on dietary intervention. In this issue of the JCI, Le Dudal and colleagues demonstrate that the lactate dehydrogenase 5 inhibitor (LDH5) stiripentol reduces urinary oxalate excretion. Importantly, stiripentol treatment of a single individual with primary hyperoxaluria reduced the urinary oxalate excretion. Together, these results support further evaluation of LDH5 as a therapeutic target for hyperoxaluria.


Asunto(s)
Oxalato de Calcio , Hiperoxaluria , Dioxolanos , Glicoles de Etileno , Humanos , Lactato Deshidrogenasa 5
4.
J Clin Invest ; 129(6): 2374-2389, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30912765

RESUMEN

Acute kidney injury (AKI) can lead to chronic kidney disease (CKD) if injury is severe and/or repair is incomplete. However, the pathogenesis of CKD following renal ischemic injury is not fully understood. Capillary rarefaction and tubular hypoxia are common findings during the AKI to CKD transition. We investigated the tubular stress response to hypoxia and demonstrated that a stress responsive transcription factor, FoxO3, was regulated by prolyl hydroxylase. Hypoxia inhibited FoxO3 prolyl hydroxylation and FoxO3 degradation, thus leading to FoxO3 accumulation and activation in tubular cells. Hypoxia-activated Hif-1α contributed to FoxO3 activation and functioned to protect kidneys, as tubular deletion of Hif-1α decreased hypoxia-induced FoxO3 activation, and resulted in more severe tubular injury and interstitial fibrosis following ischemic injury. Strikingly, tubular deletion of FoxO3 during the AKI to CKD transition aggravated renal structural and functional damage leading to a more profound CKD phenotype. We showed that tubular deletion of FoxO3 resulted in decreased autophagic response and increased oxidative injury, which may explain renal protection by FoxO3. Our study indicates that in the hypoxic kidney, stress responsive transcription factors can be activated for adaptions to counteract hypoxic insults, thus attenuating CKD development.


Asunto(s)
Muerte Celular Autofágica , Proteína Forkhead Box O3/metabolismo , Túbulos Renales/metabolismo , Estrés Oxidativo , Insuficiencia Renal Crónica/prevención & control , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Hipoxia de la Célula/genética , Fibrosis , Proteína Forkhead Box O3/genética , Túbulos Renales/patología , Ratones , Ratones Transgénicos , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
5.
Elife ; 62017 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-28577314

RESUMEN

Although most nephron segments contain one type of epithelial cell, the collecting ducts consists of at least two: intercalated (IC) and principal (PC) cells, which regulate acid-base and salt-water homeostasis, respectively. In adult kidneys, these cells are organized in rosettes suggesting functional interactions. Genetic studies in mouse revealed that transcription factor Tfcp2l1 coordinates IC and PC development. Tfcp2l1 induces the expression of IC specific genes, including specific H+-ATPase subunits and Jag1. Jag1 in turn, initiates Notch signaling in PCs but inhibits Notch signaling in ICs. Tfcp2l1 inactivation deletes ICs, whereas Jag1 inactivation results in the forfeiture of discrete IC and PC identities. Thus, Tfcp2l1 is a critical regulator of IC-PC patterning, acting cell-autonomously in ICs, and non-cell-autonomously in PCs. As a result, Tfcp2l1 regulates the diversification of cell types which is the central characteristic of 'salt and pepper' epithelia and distinguishes the collecting duct from all other nephron segments.


Asunto(s)
Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Túbulos Renales Colectores/embriología , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Ratones
6.
Stem Cell Reports ; 6(5): 757-771, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27117784

RESUMEN

To determine whether adult kidney papillary label-retaining cells (pLRCs) are specialized precursors, we analyzed their transcription profile. Among genes overexpressed in pLRCs, we selected candidate genes to perform qPCR and immunodetection of their encoded proteins. We found that Zfyve27, which encodes protrudin, identified a subpopulation of pLRCs. With Zfyve27-CreERT2 transgenic and reporter mice we generated bitransgenic animals and performed cell-lineage analysis. Post tamoxifen, Zfyve27-CreERT2 marked cells preferentially located in the upper part of the papilla. These cells were low cycling and did not generate progeny even after long-term observation, thus they did not appear to contribute to kidney homeostasis. However, after kidney injury, but only if severe, they activated a program of proliferation, migration, and morphogenesis generating multiple and long tubular segments. Remarkably these regenerated tubules were located preferentially in the kidney medulla, indicating that repair of injury in the kidney is regionally specified. These results suggest that different parts of the kidney have different progenitor cell pools.


Asunto(s)
Diferenciación Celular/genética , Médula Renal/metabolismo , Riñón/metabolismo , Regeneración/genética , Proteínas de Transporte Vesicular/genética , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Riñón/crecimiento & desarrollo , Riñón/patología , Médula Renal/crecimiento & desarrollo , Médula Renal/patología , Ratones , Células Madre/metabolismo , Tamoxifeno/farmacología , Proteínas de Transporte Vesicular/metabolismo
7.
J Clin Invest ; 125(12): 4365-74, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26517693

RESUMEN

The nephron cortical collecting duct (CCD) is composed of principal cells, which mediate Na, K, and water transport, and intercalated cells (ICs), which are specialized for acid-base transport. There are two canonical IC forms: acid-secreting α-ICs and HCO3-secreting ß-ICs. Chronic acidosis increases α-ICs at the expense of ß-ICs, thereby increasing net acid secretion by the CCD. We found by growth factor quantitative PCR array that acidosis increases expression of mRNA encoding SDF1 (or CXCL12) in kidney cortex and isolated CCDs from mouse and rabbit kidney cortex. Exogenous SDF1 or pH 6.8 media increased H+ secretion and decreased HCO3 secretion in isolated perfused rabbit CCDs. Acid-dependent changes in H+ and HCO3 secretion were largely blunted by AMD3100, which selectively blocks the SDF1 receptor CXCR4. In mice, diet-induced chronic acidosis increased α-ICs and decreased ß-ICs. Additionally, IC-specific Cxcr4 deletion prevented IC subtype alterations and magnified metabolic acidosis. SDF1 was transcriptionally regulated and a target of the hypoxia-sensing transcription factor HIF1α. IC-specific deletion of Hif1a produced no effect on mice fed an acid diet, as α-ICs increased and ß-ICs decreased similarly to that observed in WT littermates. However, Hif1a deletion in all CCD cells prevented acidosis-induced IC subtype distribution, resulting in more severe acidosis. Cultured principal cells exhibited an HIF1α-dependent increase of Sdf1 transcription in response to media acidification. Thus, our results indicate that principal cells respond to acid by producing SDF1, which then acts on adjacent ICs.


Asunto(s)
Quimiocina CXCL12/biosíntesis , Glomérulos Renales/metabolismo , Túbulos Renales Colectores/metabolismo , Animales , Células Cultivadas , Quimiocina CXCL12/genética , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Transporte Iónico/fisiología , Glomérulos Renales/citología , Túbulos Renales Colectores/citología , Ratones , Ratones Transgénicos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Conejos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
8.
J Clin Invest ; 125(8): 3304, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26237046
9.
J Clin Invest ; 125(6): 2267-70, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25985272

RESUMEN

The kidney, like other organs, grows in constant proportion to the rest of the body. When one kidney is removed, the remaining one hypertrophies. In a comprehensive series of studies, Chen et al. show that growth during maturation is mediated by the mTORC1 signaling pathway, which is induced by EGF-like peptides, and requires PI3K, PDK, AKT, mTORC2, and activation of mTORC1 through the combined effects of TSC and RHEB as part of a multiprotein complex localized on lysosomes. However, compensatory growth is mediated by amino acids, which act on mTORC1 independently of the previous pathway, and requires a class III PI3K (VPS34) that is known to be involved in vesicle trafficking to the lysosomes.


Asunto(s)
Enfermedades Renales/enzimología , Enfermedades Renales/patología , Riñón/enzimología , Riñón/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales
10.
J Am Soc Nephrol ; 26(10): 2414-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25644110

RESUMEN

The kidney developmental program encodes the intricate branching and organization of approximately 1 million functional units (nephrons). Branching regulation is poorly understood, as is the source of a 10-fold variation in nephron number. Notably, low nephron count increases the risk for developing hypertension and renal failure. To better understand the source of this variation, we analyzed the complete gestational trajectory of mouse kidney development. We constructed a computerized architectural map of the branching process throughout fetal life and found that organogenesis is composed of two distinct developmental phases, each with stage-specific rate and morphologic parameters. The early phase is characterized by a rapid acceleration in branching rate and by branching divisions that repeat with relatively reproducible morphology. The latter phase, however, is notable for a significantly decreased yet constant branching rate and the presence of nonstereotyped branching events that generate progressive variability in tree morphology until birth. Our map identifies and quantitates the contribution of four developmental mechanisms that guide organogenesis: growth, patterning, branching rate, and nephron induction. When applied to organs that developed under conditions of malnutrition or in the setting of growth factor mutation, our normative map provided an essential link between kidney architecture and the fundamental morphogenetic mechanisms that guide development. This morphogenetic map is expected to find widespread applications and help identify modifiable targets to prevent developmental programming of common diseases.


Asunto(s)
Riñón/embriología , Organogénesis , Animales , Ratones , Nefronas/embriología , Organogénesis/fisiología
11.
J Clin Invest ; 124(7): 2963-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24937428

RESUMEN

α-Intercalated cells (A-ICs) within the collecting duct of the kidney are critical for acid-base homeostasis. Here, we have shown that A-ICs also serve as both sentinels and effectors in the defense against urinary infections. In a murine urinary tract infection model, A-ICs bound uropathogenic E. coli and responded by acidifying the urine and secreting the bacteriostatic protein lipocalin 2 (LCN2; also known as NGAL). A-IC-dependent LCN2 secretion required TLR4, as mice expressing an LPS-insensitive form of TLR4 expressed reduced levels of LCN2. The presence of LCN2 in urine was both necessary and sufficient to control the urinary tract infection through iron sequestration, even in the harsh condition of urine acidification. In mice lacking A-ICs, both urinary LCN2 and urinary acidification were reduced, and consequently bacterial clearance was limited. Together these results indicate that A-ICs, which are known to regulate acid-base metabolism, are also critical for urinary defense against pathogenic bacteria. They respond to both cystitis and pyelonephritis by delivering bacteriostatic chemical agents to the lower urinary system.


Asunto(s)
Proteínas de Fase Aguda/orina , Infecciones por Escherichia coli/prevención & control , Túbulos Renales Colectores/metabolismo , Lipocalinas/orina , Proteínas Oncogénicas/orina , Proteínas Proto-Oncogénicas/orina , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena , Equilibrio Ácido-Base , Proteínas de Fase Aguda/deficiencia , Proteínas de Fase Aguda/genética , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/orina , Femenino , Humanos , Concentración de Iones de Hidrógeno , Hierro/metabolismo , Túbulos Renales Colectores/patología , Lipocalina 2 , Lipocalinas/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Receptor Toll-Like 4/metabolismo , Infecciones Urinarias/microbiología , Infecciones Urinarias/orina
12.
Nat Rev Nephrol ; 9(12): 712-3, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24189652

RESUMEN

Acidosis affects sodium and potassium excretion, likely via the pH sensitivity of ion transporters. A recent paper shows that ß-intercalated cells with deleted H(+)-ATPase release ATP into urine, which induces the production of prostaglandin E2 (PGE2). PGE2 then reduces sodium absorption in the principal cells of the cortical collecting tubule and increases potassium secretion.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Potasio en la Dieta/sangre , Sodio en la Dieta/sangre , Equilibrio Hidroelectrolítico , Animales
13.
N Engl J Med ; 369(7): 621-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23862974

RESUMEN

BACKGROUND: Congenital abnormalities of the kidney and the urinary tract are the most common cause of pediatric kidney failure. These disorders are highly heterogeneous, and the etiologic factors are poorly understood. METHODS: We performed genomewide linkage analysis and whole-exome sequencing in a family with an autosomal dominant form of congenital abnormalities of the kidney or urinary tract (seven affected family members). We also performed a sequence analysis in 311 unrelated patients, as well as histologic and functional studies. RESULTS: Linkage analysis identified five regions of the genome that were shared among all affected family members. Exome sequencing identified a single, rare, deleterious variant within these linkage intervals, a heterozygous splice-site mutation in the dual serine-threonine and tyrosine protein kinase gene (DSTYK). This variant, which resulted in aberrant splicing of messenger RNA, was present in all affected family members. Additional, independent DSTYK mutations, including nonsense and splice-site mutations, were detected in 7 of 311 unrelated patients. DSTYK is highly expressed in the maturing epithelia of all major organs, localizing to cell membranes. Knockdown in zebrafish resulted in developmental defects in multiple organs, which suggested loss of fibroblast growth factor (FGF) signaling. Consistent with this finding is the observation that DSTYK colocalizes with FGF receptors in the ureteric bud and metanephric mesenchyme. DSTYK knockdown in human embryonic kidney cells inhibited FGF-stimulated phosphorylation of extracellular-signal-regulated kinase (ERK), the principal signal downstream of receptor tyrosine kinases. CONCLUSIONS: We detected independent DSTYK mutations in 2.3% of patients with congenital abnormalities of the kidney or urinary tract, a finding that suggests that DSTYK is a major determinant of human urinary tract development, downstream of FGF signaling. (Funded by the National Institutes of Health and others.).


Asunto(s)
Mutación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Sistema Urinario/anomalías , Anomalías Urogenitales/genética , Adulto , Animales , Secuencia de Bases , Niño , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Lactante , Riñón/anomalías , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , ARN Interferente Pequeño , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Sistema Urinario/crecimiento & desarrollo , Sistema Urinario/metabolismo , Adulto Joven
14.
FEBS Lett ; 587(13): 1911-4, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23684635

RESUMEN

The intercalated cell of the collecting tubule of the mammalian kidney is specialized for the transport of H(+) and HCO3. They exist in two forms; one specialized for acid secretion and the other secretes HCO3 into the urine. We discovered many years ago that feeding animals an acid diet converts the HCO3 secreting form to an acid secreting type. Here I discuss the molecular basis of this transformation. The conversion of the cell types is mediated by an extracellular matrix protein hensin (also known as DMBT1). However much remains to be identified in the differentiation of these cells.


Asunto(s)
Túbulos Renales Colectores/citología , Células Madre Adultas/fisiología , Animales , Bicarbonatos/metabolismo , Diferenciación Celular , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Riñón/citología , Túbulos Renales Colectores/metabolismo
16.
J Clin Invest ; 122(12): 4330-3, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23187132

RESUMEN

In the kidney, the slit diaphragm joins adjacent podocytes, forming an epithelial barrier that filters plasma into the urinary space, yet retains blood cells and proteins within the circulation. In this issue of the JCI, Soda et al. have identified clathrin-mediated endocytosis as a central mechanism by which the function and structural integrity of the slit diaphragm are maintained.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Dinamina II/genética , Dinamina I/genética , Barrera de Filtración Glomerular/patología , Proteínas del Tejido Nervioso/genética , Monoéster Fosfórico Hidrolasas/genética , Podocitos/metabolismo , Animales
18.
Am J Physiol Renal Physiol ; 302(11): F1362-73, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22461304

RESUMEN

The adult kidney contains a population of low-cycling cells that resides in the papilla. These cells retain for long periods S-phase markers given as a short pulse early in life; i.e., they are label-retaining cells (LRC). In previous studies in adult rat and mice, we found that shortly after acute kidney injury many of the quiescent papillary LRC started proliferating (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009; Oliver JA, Maarouf O, Cheema FH, Martens TP, Al-Awqati Q. J Clin Invest 114: 795-804, 2004) and, with cell-tracking experiments, we found upward migration of some papillary cells including LRC (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009). To identify molecular cues involved in the activation (i.e., proliferation and/or migration) of the papillary LRC that follows injury, we isolated these cells from the H2B-GFP mice and found that they migrated and proliferated in response to the cytokine stromal cell-derived factor-1 (SDF-1). Moreover, in a papillary organ culture assay, the cell growth out of the upper papilla was dependent on the interaction of SDF-1 with its receptor Cxcr4. Interestingly, location of these two proteins in the kidney revealed a complementary location, with SDF-1 being preferentially expressed in the medulla and Cxcr4 more abundant in the papilla. Blockade of Cxcr4 in vivo prevented mobilization of papillary LRC after transient kidney ischemic injury and worsened its functional consequences. The data indicate that the SDF-1/Cxcr4 axis is a critical regulator of papillary LRC activation following transient kidney injury and during organ repair.


Asunto(s)
Lesión Renal Aguda/patología , Quimiocina CXCL12/farmacología , Enfermedades Renales/patología , Médula Renal/crecimiento & desarrollo , Lesión Renal Aguda/fisiopatología , Animales , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Separación Celular , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Femenino , Inmunohistoquímica , Indicadores y Reactivos , Enfermedades Renales/fisiopatología , Médula Renal/patología , Médula Renal/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo
20.
Physiology (Bethesda) ; 26(4): 266-72, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21841074

RESUMEN

The intercalated cell of collecting ducts of the kidney is of two forms, the α form secretes acid, whereas the ß form secretes HCO(3). Here, we review recent work that shows that the α form is derived from the ß form and that the pathway is mediated by an extracellular matrix protein called hensin/DMBT1.


Asunto(s)
Diferenciación Celular/fisiología , Riñón/citología , Animales , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Riñón/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA