Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 321(6): F689-F704, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34693742

RESUMEN

Macula densa (MD) cells, a chief sensory cell type in the nephron, are endowed with unique microanatomic features including a high density of protein synthetic organelles and secretory vesicles in basal cell processes ("maculapodia") that suggest a so far unknown high rate of MD protein synthesis. This study aimed to explore the rate and regulation of MD protein synthesis and their effects on glomerular function using novel transgenic mouse models, newly established fluorescence cell biology techniques, and intravital microscopy. Sox2-tdTomato kidney tissue sections and an O-propargyl puromycin incorporation-based fluorescence imaging assay showed that MD cells have the highest level of protein synthesis within the kidney cortex followed by intercalated cells and podocytes. Genetic gain of function of mammalian target of rapamycin (mTOR) signaling specifically in MD cells (in MD-mTORgof mice) or their physiological activation by low-salt diet resulted in further significant increases in the synthesis of MD proteins. Specifically, these included both classic and recently identified MD-specific proteins such as cyclooxygenase 2, microsomal prostaglandin E2 synthase 1, and pappalysin 2. Intravital imaging of the kidney using multiphoton microscopy showed significant increases in afferent and efferent arteriole and glomerular capillary diameters and blood flow in MD-mTORgof mice coupled with an elevated glomerular filtration rate. The presently identified high rate of MD protein synthesis that is regulated by mTOR signaling is a novel component of the physiological activation and glomerular hemodynamic regulatory functions of MD cells that remains to be fully characterized.NEW & NOTEWORTHY This study discovered the high rate of protein synthesis in macula densa (MD) cells by applying direct imaging techniques with single cell resolution. Physiological activation and mammalian target of rapamycin signaling played important regulatory roles in this process. This new feature is a novel component of the tubuloglomerular cross talk and glomerular hemodynamic regulatory functions of MD cells. Future work is needed to elucidate the nature and (patho)physiological role of the specific proteins synthesized by MD cells.


Asunto(s)
Aparato Yuxtaglomerular/metabolismo , Biosíntesis de Proteínas , Animales , Comunicación Autocrina , Dieta Hiposódica , Tasa de Filtración Glomerular , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Intravital , Aparato Yuxtaglomerular/citología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Comunicación Paracrina , Renina/metabolismo , Transducción de Señal , Sodio en la Dieta/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína Fluorescente Roja
2.
Am J Physiol Renal Physiol ; 320(3): F492-F504, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33491562

RESUMEN

Although macula densa (MD) cells are chief regulatory cells in the nephron with unique microanatomical features, they have been difficult to study in full detail due to their inaccessibility and limitations in earlier microscopy techniques. The present study used a new mouse model with a comprehensive imaging approach to visualize so far unexplored microanatomical features of MD cells, their regulation, and functional relevance. MD-GFP mice with conditional and partial induction of green fluorescent protein (GFP) expression, which specifically and intensely illuminated only single MD cells, were used with fluorescence microscopy of fixed tissue and live MD cells in vitro and in vivo with complementary electron microscopy of the rat, rabbit, and human kidney. An elaborate network of major and minor cell processes, here named maculapodia, were found at the cell base, projecting toward other MD cells and the glomerular vascular pole. The extent of maculapodia showed upregulation by low dietary salt intake and the female sex. Time-lapse imaging of maculapodia revealed highly dynamic features including rapid outgrowth and an extensive vesicular transport system. Electron microscopy of rat, rabbit, and human kidneys and three-dimensional volume reconstruction in optically cleared whole-mount MD-GFP mouse kidneys further confirmed the presence and projections of maculapodia into the extraglomerular mesangium and afferent and efferent arterioles. The newly identified dynamic and secretory features of MD cells suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD cells and between MD and other target cells.NEW & NOTEWORTHY This study illuminated a physiologically regulated dense network of basal cell major and minor processes (maculapodia) in macula densa (MD) cells. The newly identified dynamic and secretory features of these microanatomical structures suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD and other target cells. Detailed characterization of the function and molecular details of MD cell intercellular communications and their role in physiology and disease warrant further studies.


Asunto(s)
Mesangio Glomerular/ultraestructura , Aparato Yuxtaglomerular/ultraestructura , Glomérulos Renales/ultraestructura , Túbulos Renales/ultraestructura , Animales , Comunicación Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/ultraestructura , Mesangio Glomerular/citología , Glomérulos Renales/citología , Túbulos Renales/citología , Ratones , Conejos , Ratas
3.
J Am Soc Nephrol ; 31(7): 1555-1568, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32487560

RESUMEN

BACKGROUND: The physiologic role of renomedullary interstitial cells, which are uniquely and abundantly found in the renal inner medulla, is largely unknown. Endothelin A receptors regulate multiple aspects of renomedullary interstitial cell function in vitro. METHODS: To assess the effect of targeting renomedullary interstitial cell endothelin A receptors in vivo, we generated a mouse knockout model with inducible disruption of renomedullary interstitial cell endothelin A receptors at 3 months of age. RESULTS: BP and renal function were similar between endothelin A receptor knockout and control mice during normal and reduced sodium or water intake. In contrast, on a high-salt diet, compared with control mice, the knockout mice had reduced BP; increased urinary sodium, potassium, water, and endothelin-1 excretion; increased urinary nitrite/nitrate excretion associated with increased noncollecting duct nitric oxide synthase-1 expression; increased PGE2 excretion associated with increased collecting duct cyclooxygenase-1 expression; and reduced inner medullary epithelial sodium channel expression. Water-loaded endothelin A receptor knockout mice, compared with control mice, had markedly enhanced urine volume and reduced urine osmolality associated with increased urinary endothelin-1 and PGE2 excretion, increased cyclooxygenase-2 protein expression, and decreased inner medullary aquaporin-2 protein content. No evidence of endothelin-1-induced renomedullary interstitial cell contraction was observed. CONCLUSIONS: Disruption of renomedullary interstitial cell endothelin A receptors reduces BP and increases salt and water excretion associated with enhanced production of intrinsic renal natriuretic and diuretic factors. These studies indicate that renomedullary interstitial cells can modulate BP and renal function under physiologic conditions.


Asunto(s)
Presión Sanguínea , Médula Renal/fisiología , Receptor de Endotelina A/fisiología , Aldosterona/sangre , Animales , Arginina Vasopresina/orina , Calcio/metabolismo , Diuresis/efectos de los fármacos , Endotelina-1/farmacología , Endotelina-1/orina , Canales Epiteliales de Sodio/metabolismo , Femenino , Genotipo , Tasa de Filtración Glomerular , Ácido Hialurónico/metabolismo , Médula Renal/citología , Médula Renal/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Natriuresis/efectos de los fármacos , Nitratos/orina , Nitritos/orina , Potasio/orina , ARN Mensajero/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Sodio/orina , Cloruro de Sodio Dietético/administración & dosificación , Tamoxifeno/farmacología , Agua/administración & dosificación , Agua/metabolismo
4.
Annu Rev Physiol ; 78: 391-414, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26667077

RESUMEN

To maintain metabolic homeostasis, the body must be able to monitor the concentration of a large number of substances, including metabolites, in real time and to use that information to regulate the activities of different metabolic pathways. Such regulation is achieved by the presence of sensors, termed metabolite receptors, in various tissues and cells of the body, which in turn convey the information to appropriate regulatory or positive or negative feedback systems. In this review, we cover the unique roles of metabolite receptors in renal and vascular function. These receptors play a wide variety of important roles in maintaining various aspects of homeostasis-from salt and water balance to metabolism-by sensing metabolites from a wide variety of sources. We discuss the role of metabolite sensors in sensing metabolites generated locally, metabolites generated at distant tissues or organs, or even metabolites generated by resident microbes. Metabolite receptors are also involved in various pathophysiological conditions and are being recognized as potential targets for new drugs. By highlighting three receptor families-(a) citric acid cycle intermediate receptors, (b) purinergic receptors, and


Asunto(s)
Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiología , Homeostasis/fisiología , Riñón/metabolismo , Riñón/fisiología , Receptores de Superficie Celular/metabolismo , Animales , Humanos
5.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29123029

RESUMEN

Normal renin synthesis and secretion is important for the maintenance of juxtaglomerular apparatus architecture. Mice lacking a functional Ren1d gene are devoid of renal juxtaglomerular cell granules and exhibit an altered macula densa morphology. Due to the species-specificity of renin activity, transgenic mice are ideal models for experimentally investigating and manipulating expression patterns of the human renin gene in a native cellular environment without confounding renin-angiotensin system interactions. A 55-kb transgene encompassing the human renin locus was crossed onto the mouse Ren1d-null background, restoring granulation in juxtaglomerular cells. Correct processing of human renin in dense core granules was confirmed by immunogold labeling. After stimulation of the renin-angiotensin system, juxtaglomerular cells contained rhomboid protogranules with paracrystalline contents, dilated rough endoplasmic reticulum, and electron-lucent granular structures. However, complementation of Ren1d-/- mice with human renin was unable to rescue the abnormality seen in macula densa structure. The juxtaglomerular apparatus was still able to respond to tubuloglomerular feedback in isolated perfused juxtaglomerular apparatus preparations, although minor differences in glomerular tuft contractility and macula densa cell calcium handling were observed. This study reveals that the human renin protein is able to complement the mouse Ren1d-/- non-granulated defect and suggests that granulopoiesis requires a structural motif that is conserved between the mouse Ren1d and human renin proteins. It also suggests that the altered macula densa phenotype is related to the activity of the renin-1d enzyme in a local juxtaglomerular renin-angiotensin system.


Asunto(s)
Prueba de Complementación Genética , Aparato Yuxtaglomerular/enzimología , Renina/biosíntesis , Transgenes , Animales , Humanos , Aparato Yuxtaglomerular/patología , Ratones , Ratones Noqueados , Renina/genética
6.
Kidney Int ; 95(1): 94-107, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389198

RESUMEN

Aldosterone contributes to end-organ damage in heart failure and chronic kidney disease. Mineralocorticoid-receptor inhibitors limit activation of the receptor by aldosterone and slow disease progression, but side effects, including hyperkalemia, limit their clinical use. Damage to the endothelial glycocalyx (a luminal biopolymer layer) has been implicated in the pathogenesis of endothelial dysfunction and albuminuria, but to date no one has investigated whether the glomerular endothelial glycocalyx is affected by aldosterone. In vitro, human glomerular endothelial cells exposed to 0.1 nM aldosterone and 145 mMol NaCl exhibited reduced cell surface glycocalyx components (heparan sulfate and syndecan-4) and disrupted shear sensing consistent with damage of the glycocalyx. In vivo, administration of 0.6 µg/g/d of aldosterone (subcutaneous minipump) and 1% NaCl drinking water increased glomerular matrix metalloproteinase 2 activity, reduced syndecan 4 expression, and caused albuminuria. Intravital multiphoton imaging confirmed that aldosterone caused damage of the glomerular endothelial glycocalyx and increased the glomerular sieving coefficient for albumin. Targeting matrix metalloproteinases 2 and 9 with a specific gelatinase inhibitor preserved the glycocalyx, blocked the rise in glomerular sieving coefficient, and prevented albuminuria. Together these data suggest that preservation of the glomerular endothelial glycocalyx may represent a novel strategy for limiting the pathological effects of aldosterone.


Asunto(s)
Albuminuria/patología , Aldosterona/metabolismo , Glicocálix/patología , Insuficiencia Renal Crónica/patología , Albuminuria/orina , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Glicocálix/efectos de los fármacos , Heparitina Sulfato/metabolismo , Humanos , Glomérulos Renales/citología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Insuficiencia Renal Crónica/orina , Cloruro de Sodio/farmacología , Sindecano-4/metabolismo
7.
Circ Res ; 120(9): 1426-1439, 2017 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-28167653

RESUMEN

RATIONALE: Lymphatic vessels function to drain interstitial fluid from a variety of tissues. Although shear stress generated by fluid flow is known to trigger lymphatic expansion and remodeling, the molecular basis underlying flow-induced lymphatic growth is unknown. OBJECTIVE: We aimed to gain a better understanding of the mechanism by which laminar shear stress activates lymphatic proliferation. METHODS AND RESULTS: Primary endothelial cells from dermal blood and lymphatic vessels (blood vascular endothelial cells and lymphatic endothelial cells [LECs]) were exposed to low-rate steady laminar flow. Shear stress-induced molecular and cellular responses were defined and verified using various mutant mouse models. Steady laminar flow induced the classic shear stress responses commonly in blood vascular endothelial cells and LECs. Surprisingly, however, only LECs showed enhanced cell proliferation by regulating the vascular endothelial growth factor (VEGF)-A, VEGF-C, FGFR3, and p57/CDKN1C genes. As an early signal mediator, ORAI1, a pore subunit of the calcium release-activated calcium channel, was identified to induce the shear stress phenotypes and cell proliferation in LECs responding to the fluid flow. Mechanistically, ORAI1 induced upregulation of Krüppel-like factor (KLF)-2 and KLF4 in the flow-activated LECs, and the 2 KLF proteins cooperate to regulate VEGF-A, VEGF-C, FGFR3, and p57 by binding to the regulatory regions of the genes. Consistently, freshly isolated LECs from Orai1 knockout embryos displayed reduced expression of KLF2, KLF4, VEGF-A, VEGF-C, and FGFR3 and elevated expression of p57. Accordingly, mouse embryos deficient in Orai1, Klf2, or Klf4 showed a significantly reduced lymphatic density and impaired lymphatic development. CONCLUSIONS: Our study identified a molecular mechanism for laminar flow-activated LEC proliferation.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Linfangiogénesis , Mecanotransducción Celular , Proteína ORAI1/metabolismo , Animales , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Endotelio Linfático/patología , Endotelio Linfático/fisiopatología , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Genotipo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Ratones Noqueados , Proteína ORAI1/deficiencia , Proteína ORAI1/genética , Fenotipo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Estrés Mecánico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo
8.
Am J Physiol Renal Physiol ; 315(3): F521-F534, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29667908

RESUMEN

The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.


Asunto(s)
Presión Sanguínea , Aparato Yuxtaglomerular/enzimología , ATPasas de Translocación de Protón/metabolismo , Receptores de Superficie Celular/metabolismo , Sistema Renina-Angiotensina , Renina/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Técnicas Biosensibles , Presión Sanguínea/efectos de los fármacos , Captopril/farmacología , Ciclooxigenasa 2/metabolismo , Dieta Hiposódica , Dinoprostona/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Aparato Yuxtaglomerular/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandina-E Sintasas/metabolismo , ATPasas de Translocación de Protón/deficiencia , ATPasas de Translocación de Protón/genética , Ratas Sprague-Dawley , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Sistema Renina-Angiotensina/efectos de los fármacos , Vías Secretoras , Transducción de Señal , ATPasas de Translocación de Protón Vacuolares/genética , Receptor de Prorenina
9.
Pflugers Arch ; 469(7-8): 965-974, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28664407

RESUMEN

The development of podocyte injury and albuminuria in various glomerular pathologies is still incompletely understood due to technical limitations in studying the glomerular filtration barrier (GFB) in real-time. We aimed to directly visualize the early morphological and functional changes of the GFB during the development of focal segmental glomerulosclerosis (FSGS) using a combination of transmission electron microscopy (TEM) and in vivo multiphoton microscopy (MPM) in the rat puromycin aminonucleoside (PAN) model. We hypothesized that this combined TEM + MPM experimental approach would provide a major technical improvement that would benefit our mechanistic understanding of podocyte detachment. Male Sprague-Dawley (for TEM) or Munich-Wistar-Frömter (for MPM) rats were given a single dose of 100-150 mg/kg body weight PAN i.p. and were either sacrificed and the kidneys processed for TEM or surgically instrumented for in vivo MPM imaging at various times 2-14 days after PAN administration. Both techniques demonstrated hypertrophy and cystic dilatations of the subpodocyte space that developed as early as 2-3 days after PAN. Adhesions of the visceral epithelium to the parietal Bowman's capsule (synechiae) appeared at days 8-10. TEM provided unmatched resolution of podocyte foot process remodeling, while MPM revealed the rapid dynamics of pseudocyst filling, emptying, and rupture, as well as endothelial and podocyte injury, misdirected filtration, and podocyte shedding. Due to the complementary advantages of TEM and MPM, this combined approach can provide an unusally comprehensive and dynamic portrayal of the alterations in podocyte morphology and function during FSGS development. The results advance our understanding of the role and importance of the various cell types, hemodynamics, and mechanical forces in the development of glomerular pathology.


Asunto(s)
Movimiento Celular , Glomerulonefritis/patología , Podocitos/ultraestructura , Animales , Glomerulonefritis/etiología , Masculino , Podocitos/fisiología , Puromicina Aminonucleósido/toxicidad , Ratas , Ratas Sprague-Dawley , Ratas Wistar
10.
Purinergic Signal ; 13(2): 239-248, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28233082

RESUMEN

Previously, we localized ADP-activated P2Y12 receptor (R) in rodent kidney and showed that its blockade by clopidogrel bisulfate (CLPD) attenuates lithium (Li)-induced nephrogenic diabetes insipidus (NDI). Here, we evaluated the effect of prasugrel (PRSG) administration on Li-induced NDI in mice. Both CLPD and PRSG belong to the thienopyridine class of ADP receptor antagonists. Groups of age-matched adult male B6D2 mice (N = 5/group) were fed either regular rodent chow (CNT), or with added LiCl (40 mmol/kg chow) or PRSG in drinking water (10 mg/kg bw/day) or a combination of LiCl and PRSG for 14 days and then euthanized. Water intake and urine output were determined and blood and kidney tissues were collected and analyzed. PRSG administration completely suppressed Li-induced polydipsia and polyuria and significantly prevented Li-induced decreases in AQP2 protein abundance in renal cortex and medulla. However, PRSG either alone or in combination with Li did not have a significant effect on the protein abundances of NKCC2 or NCC in the cortex and/or medulla. Immunofluorescence microscopy revealed that PRSG administration prevented Li-induced alterations in cellular disposition of AQP2 protein in medullary collecting ducts. Serum Li, Na, and osmolality were not affected by the administration of PRSG. Similar to CLPD, PRSG administration had no effect on Li-induced increase in urinary Na excretion. However, unlike CLPD, PRSG did not augment Li-induced increase in urinary arginine vasopressin (AVP) excretion. Taken together, these data suggest that the pharmacological inhibition of P2Y12-R by the thienopyridine group of drugs may potentially offer therapeutic benefits in Li-induced NDI.


Asunto(s)
Diabetes Insípida Nefrogénica/inducido químicamente , Riñón/efectos de los fármacos , Cloruro de Litio/toxicidad , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Animales , Masculino , Ratones
11.
Am J Physiol Renal Physiol ; 311(6): F1230-F1242, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27335372

RESUMEN

Pericytes are tissue-resident mesenchymal progenitor cells anatomically associated with the vasculature that have been shown to participate in tissue regeneration. Here, we tested the hypothesis that kidney pericytes, derived from FoxD1+ mesodermal progenitors during embryogenesis, are necessary for postnatal kidney homeostasis. Diphtheria toxin delivery to FoxD1Cre::RsDTR transgenic mice resulted in selective ablation of >90% of kidney pericytes but not other cell lineages. Abrupt increases in plasma creatinine, blood urea nitrogen, and albuminuria within 96 h indicated acute kidney injury in pericyte-ablated mice. Loss of pericytes led to a rapid accumulation of neutral lipid vacuoles, swollen mitochondria, and apoptosis in tubular epithelial cells. Pericyte ablation led to endothelial cell swelling, reduced expression of vascular homeostasis markers, and peritubular capillary loss. Despite the observed injury, no signs of the acute inflammatory response were observed. Pathway enrichment analysis of genes expressed in kidney pericytes in vivo identified basement membrane proteins, angiogenic factors, and factors regulating vascular tone as major regulators of vascular function. Using novel microphysiological devices, we recapitulated human kidney peritubular capillaries coated with pericytes and showed that pericytes regulate permeability, basement membrane deposition, and microvascular tone. These findings suggest that through the active support of the microvasculature, pericytes are essential to adult kidney homeostasis.


Asunto(s)
Lesión Renal Aguda/metabolismo , Capilares/metabolismo , Endotelio Vascular/metabolismo , Riñón/irrigación sanguínea , Pericitos/metabolismo , Animales , Riñón/metabolismo , Ratones , Ratones Transgénicos , Microvasos/metabolismo , Permeabilidad
12.
Kidney Int ; 90(5): 941-942, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27742198

RESUMEN

Proper histological measurement of kidney fibrosis is essentially important in both clinical pathology and basic research using animal models of chronic kidney disease (CKD). However, standard histology techniques and their blind evaluation are cumbersome. Ranjit et al. applied an advanced optical microscopy technique for hassle-free, unbiased, and highly sensitive characterization of kidney fibrosis and tested it in a classic model of chronic kidney disease in mice. This commentary emphasizes the advantages and future promise of this new approach.


Asunto(s)
Insuficiencia Renal Crónica , Microscopía de Generación del Segundo Armónico , Animales , Fibrosis , Riñón , Ratones , Microscopía
13.
Curr Opin Nephrol Hypertens ; 25(3): 168-73, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27008595

RESUMEN

PURPOSE OF REVIEW: The review aims to provide a brief summary and evaluation of the current state of research that uses multiphoton fluorescence microscopy for intravital kidney imaging. RECENT FINDINGS: Direct visualization of the glomerular filter, proximal and distal tubule segments, and the renal vasculature in the living, intact kidney in zebrafish, mouse, and rat models with high temporal and spatial resolution provided new insights into the function of the normal and diseased kidney. New technical developments in fluorescence excitation and detection, in combination with transgenic animal models for cell function and fate mapping, and serial imaging of the same glomerulus in the same animal over several days further advanced the field of nephrology research, and the understanding of disease mechanisms. SUMMARY: Intravital multiphoton imaging has solved many critical technical barriers in kidney research and allowed the dynamic portrayal of the structure and function of various renal cell types in vivo. It has become a widely used research technique, with significant past achievements, and tremendous potential for future development and applications for the study and better understanding of kidney diseases.


Asunto(s)
Microscopía Intravital , Enfermedades Renales/diagnóstico , Riñón/patología , Microscopía de Fluorescencia por Excitación Multifotónica , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Riñón/irrigación sanguínea , Enfermedades Renales/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Modelos Animales
14.
Proc Natl Acad Sci U S A ; 110(19): 7928-33, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610411

RESUMEN

The Na(+) concentration of the intracellular milieu is very low compared with the extracellular medium. Transport of Na(+) along this gradient is used to fuel secondary transport of many solutes, and thus plays a major role for most cell functions including the control of cell volume and resting membrane potential. Because of a continuous leak, Na(+) has to be permanently removed from the intracellular milieu, a process that is thought to be exclusively mediated by the Na(+)/K(+)-ATPase in animal cells. Here, we show that intercalated cells of the mouse kidney are an exception to this general rule. By an approach combining two-photon imaging of isolated renal tubules, physiological studies, and genetically engineered animals, we demonstrate that inhibition of the H(+) vacuolar-type ATPase (V-ATPase) caused drastic cell swelling and depolarization, and also inhibited the NaCl absorption pathway that we recently discovered in intercalated cells. In contrast, pharmacological blockade of the Na(+)/K(+)-ATPase had no effects. Basolateral NaCl exit from ß-intercalated cells was independent of the Na(+)/K(+)-ATPase but critically relied on the presence of the basolateral ion transporter anion exchanger 4. We conclude that not all animal cells critically rely on the sodium pump as the unique bioenergizer, but can be replaced by the H(+) V-ATPase in renal intercalated cells. This concept is likely to apply to other animal cell types characterized by plasma membrane expression of the H(+) V-ATPase.


Asunto(s)
Riñón/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Sodio/metabolismo , Absorción , Animales , Membrana Celular/metabolismo , Células Cultivadas , Antiportadores de Cloruro-Bicarbonato/genética , Inmunohistoquímica , Iones , Potenciales de la Membrana , Ratones , Ratones Noqueados , Perfusión , Bombas de Protones/fisiología , Cloruro de Sodio/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(11): 4410-5, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23401498

RESUMEN

Olfactory receptors are G protein-coupled receptors that mediate olfactory chemosensation and serve as chemosensors in other tissues. We find that Olfr78, an olfactory receptor expressed in the kidney, responds to short chain fatty acids (SCFAs). Olfr78 is expressed in the renal juxtaglomerular apparatus, where it mediates renin secretion in response to SCFAs. In addition, both Olfr78 and G protein-coupled receptor 41 (Gpr41), another SCFA receptor, are expressed in smooth muscle cells of small resistance vessels. Propionate, a SCFA shown to induce vasodilation ex vivo, produces an acute hypotensive response in wild-type mice. This effect is differentially modulated by disruption of Olfr78 and Gpr41 expression. SCFAs are end products of fermentation by the gut microbiota and are absorbed into the circulation. Antibiotic treatment reduces the biomass of the gut microbiota and elevates blood pressure in Olfr78 knockout mice. We conclude that SCFAs produced by the gut microbiota modulate blood pressure via Olfr78 and Gpr41.


Asunto(s)
Presión Sanguínea/fisiología , Intestinos/microbiología , Riñón/metabolismo , Metagenoma/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Odorantes/metabolismo , Renina/metabolismo , Transducción de Señal/fisiología , Animales , Biomasa , Presión Sanguínea/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/microbiología , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Propionatos/metabolismo , Propionatos/farmacología , Receptores Acoplados a Proteínas G/genética , Receptores Odorantes/genética , Transducción de Señal/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
16.
J Am Soc Nephrol ; 26(12): 2978-87, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25855780

RESUMEN

P2Y12 receptor (P2Y12-R) signaling is mediated through Gi, ultimately reducing cellular cAMP levels. Because cAMP is a central modulator of arginine vasopressin (AVP)-induced water transport in the renal collecting duct (CD), we hypothesized that if expressed in the CD, P2Y12-R may play a role in renal handling of water in health and in nephrogenic diabetes insipidus. We found P2Y12-R mRNA expression in rat kidney, and immunolocalized its protein and aquaporin-2 (AQP2) in CD principal cells. Administration of clopidogrel bisulfate, an irreversible inhibitor of P2Y12-R, significantly increased urine concentration and AQP2 protein in the kidneys of Sprague-Dawley rats. Notably, clopidogrel did not alter urine concentration in Brattleboro rats that lack AVP. Clopidogrel administration also significantly ameliorated lithium-induced polyuria, improved urine concentrating ability and AQP2 protein abundance, and reversed the lithium-induced increase in free-water excretion, without decreasing blood or kidney tissue lithium levels. Clopidogrel administration also augmented the lithium-induced increase in urinary AVP excretion and suppressed the lithium-induced increase in urinary nitrates/nitrites (nitric oxide production) and 8-isoprostane (oxidative stress). Furthermore, selective blockade of P2Y12-R by the reversible antagonist PSB-0739 in primary cultures of rat inner medullary CD cells potentiated the expression of AQP2 and AQP3 mRNA, and cAMP production induced by dDAVP (desmopressin). In conclusion, pharmacologic blockade of renal P2Y12-R increases urinary concentrating ability by augmenting the effect of AVP on the kidney and ameliorates lithium-induced NDI by potentiating the action of AVP on the CD. This strategy may offer a novel and effective therapy for lithium-induced NDI.


Asunto(s)
Arginina Vasopresina/metabolismo , Diabetes Insípida Nefrogénica/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiopatología , Receptores Purinérgicos P2Y12/metabolismo , Animales , Acuaporina 2/análisis , Acuaporina 2/efectos de los fármacos , Acuaporina 2/orina , Arginina Vasopresina/efectos de los fármacos , Arginina Vasopresina/orina , Clopidogrel , Desamino Arginina Vasopresina/metabolismo , Diabetes Insípida Nefrogénica/inducido químicamente , Diabetes Insípida Nefrogénica/fisiopatología , Capacidad de Concentración Renal/efectos de los fármacos , Médula Renal/química , Túbulos Renales Colectores/química , Litio , Masculino , Antagonistas del Receptor Purinérgico P2Y/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Brattleboro , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y12/análisis , Receptores Purinérgicos P2Y12/genética , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Agua/metabolismo
17.
Annu Rev Physiol ; 74: 325-49, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21888509

RESUMEN

The distal nephron plays a critical role in the renal control of homeostasis. Until very recently most studies focused on the control of Na(+), K(+), and water balance by principal cells of the collecting duct and the regulation of solute and water by hormones from the renin-angiotensin-aldosterone system and by antidiuretic hormone. However, recent studies have revealed the unexpected importance of renal intercalated cells, a subtype of cells present in the connecting tubule and collecting ducts. Such cells were thought initially to be involved exclusively in acid-base regulation. However, it is clear now that intercalated cells absorb NaCl and K(+) and hence may participate in the regulation of blood pressure and potassium balance. The second paradigm-challenging concept we highlight is the emerging importance of local paracrine factors that play a critical role in the renal control of water and electrolyte balance.


Asunto(s)
Electrólitos/metabolismo , Túbulos Renales Distales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico/fisiología , Presión Sanguínea/fisiología , Cloruros/metabolismo , Colon/metabolismo , Diuréticos/farmacología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Homeostasis/fisiología , Humanos , Calicreínas/metabolismo , Calicreínas/fisiología , Túbulos Renales Distales/citología , Potasio/metabolismo , Sistema Renina-Angiotensina/fisiología , Canales de Sodio/fisiología , Cloruro de Sodio/metabolismo , Tiazidas/farmacología
18.
Kidney Int ; 88(1): 44-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25738253

RESUMEN

Intravital imaging using multiphoton microscopy (MPM) has become an increasingly popular and widely used experimental technique in kidney research over the past few years. MPM allows deep optical sectioning of the intact, living kidney tissue with submicron resolution, which is unparalleled among intravital imaging approaches. MPM has solved a long-standing critical technical barrier in renal research to study several complex and inaccessible cell types and anatomical structures in vivo in their native environment. Comprehensive and quantitative kidney structure and function MPM studies helped our better understanding of the cellular and molecular mechanisms of the healthy and diseased kidney. This review summarizes recent in vivo MPM studies with a focus on the glomerulus and the filtration barrier, although select, glomerulus-related renal vascular and tubular functions are also mentioned. The latest applications of serial MPM of the same glomerulus in vivo, in the intact kidney over several days, during the progression of glomerular disease are discussed. This visual approach, in combination with genetically encoded fluorescent markers of cell lineage, has helped track the fate and function (e.g., cell calcium changes) of single podocytes during the development of glomerular pathologies, and provided visual proof for the highly dynamic, rather than static, nature of the glomerular environment. Future intravital imaging applications have the promise to further push the limits of optical microscopy, and to advance our understanding of the mechanisms of kidney injury. Also, MPM will help to study new mechanisms of tissue repair and regeneration, a cutting-edge area of kidney research.


Asunto(s)
Barrera de Filtración Glomerular/anatomía & histología , Barrera de Filtración Glomerular/fisiología , Microscopía Intravital/métodos , Enfermedades Renales/patología , Microscopía de Fluorescencia por Excitación Multifotónica , Animales , Calcio/metabolismo , Movimiento Celular , Enfermedades Renales/fisiopatología , Ratones , Podocitos/fisiología , Ratas , Imagen de Lapso de Tiempo , Pez Cebra
19.
Biochem Biophys Res Commun ; 457(1): 19-22, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25529452

RESUMEN

This study describes a technical breakthrough in endolymphatic sac research, made possible by the use of the recently generated Prox1-GFP transgenic mouse model. Whole-mount imaging techniques through the decalcified temporal bone and three-dimensional observations of Prox1-GFP mouse tissue revealed the positive labeling of the endolymphatic sac in adult stage, and allowed, for the first time, the GFP-based identification of endolymphatic sac epithelial cells. Prox1 expression was observed in all parts of the endolymphatic sac epithelia. In intermediate portion of the endolymphatic sac, mitochondria-rich cells did not express Prox1, although ribosome-rich cells showed strong GFP labeling. The anatomical relationship between the endolymphatic sac and the surrounding vasculature was directly observed. In the endolymphatic sac, expression of Prox1 may suggest progenitor cell-like pluripotency or developmental similarity to systemic lymphatic vessels in other organs. This whole-mount imaging technique of the endolymphatic sac can be combined with other conventional histological, sectioning, and labeling techniques and will be very useful for future endolymphatic sac research.


Asunto(s)
Saco Endolinfático/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Imagenología Tridimensional , Proteínas Supresoras de Tumor/metabolismo , Envejecimiento/metabolismo , Animales , Saco Endolinfático/citología , Células Epiteliales/metabolismo , Fluorescencia , Ratones Transgénicos
20.
Purinergic Signal ; 11(4): 507-18, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26386699

RESUMEN

Lithium (Li) administration causes deranged expression and function of renal aquaporins and sodium channels/transporters resulting in nephrogenic diabetes insipidus (NDI). Extracellular nucleotides (ATP/ADP/UTP), via P2 receptors, regulate these transport functions. We tested whether clopidogrel bisulfate (CLPD), an antagonist of ADP-activated P2Y(12) receptor, would affect Li-induced alterations in renal aquaporins and sodium channels/transporters. Adult mice were treated for 14 days with CLPD and/or Li and euthanized. Urine and kidneys were collected for analysis. When administered with Li, CLPD ameliorated polyuria, attenuated the rise in urine prostaglandin E2 (PGE2), and resulted in significantly higher urinary arginine vasopressin (AVP) and aldosterone levels as compared to Li treatment alone. However, urine sodium excretion remained elevated. Semi-quantitative immunoblotting revealed that CLPD alone increased renal aquaporin 2 (AQP2), Na-K-2Cl cotransporter (NKCC2), Na-Cl cotransporter (NCC), and the subunits of the epithelial Na channel (ENaC) in medulla by 25-130 %. When combined with Li, CLPD prevented downregulation of AQP2, Na-K-ATPase, and NKCC2 but was less effective against downregulation of cortical α- or γ-ENaC (70 kDa band). Thus, CLPD primarily attenuated Li-induced downregulation of proteins involved in water conservation (AVP-sensitive), with modest effects on aldosterone-sensitive proteins potentially explaining sustained natriuresis. Confocal immunofluorescence microscopy revealed strong labeling for P2Y(12)-R in proximal tubule brush border and blood vessels in the cortex and less intense labeling in medullary thick ascending limb and the collecting ducts. Therefore, there is the potential for CLPD to be directly acting at the tubule sites to mediate these effects. In conclusion, P2Y(12)-R may represent a novel therapeutic target for Li-induced NDI.


Asunto(s)
Agua Corporal/metabolismo , Riñón/metabolismo , Litio/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Canales de Sodio/metabolismo , Ticlopidina/análogos & derivados , Aldosterona/orina , Animales , Acuaporina 2/metabolismo , Acuaporinas/metabolismo , Arginina Vasopresina/orina , Clopidogrel , Dinoprostona/orina , Canales Epiteliales de Sodio/metabolismo , Riñón/efectos de los fármacos , Masculino , Ratones , Poliuria/inducido químicamente , Poliuria/tratamiento farmacológico , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/metabolismo , Canales de Sodio/efectos de los fármacos , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Ticlopidina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA