Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 726
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Adv Physiol Educ ; 47(3): 665-671, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37439318

RESUMEN

Countercurrent multiplication (CCM) is widely accepted as the mechanism for the generation of the corticopapillary osmotic gradient in the outer medulla of mammalian kidneys. However, several issues in the literature cause the current explanations of CCM to be inefficient and incomplete. As a result, it is challenging to clearly explain CCM in physiology education. The goal of this article is to share a modified version of CCM with more understandable explanation in the hopes of motivating peer discussion, further improvement, and future research. To reach this goal, the logical processes leading to CCM are first analyzed, which results in a set of formulas that serve as the principles governing CCM. Next, the cessation of CCM is addressed to provide a complete picture of the modified version of CCM. Throughout these two steps, the issues mentioned above are identified and addressed so that how the modified version of CCM eliminates these issues becomes clear. The formulas mentioned above are provided in the Tables S1, S2, and S3 (all Supplemental material is available in the Supplemental Excel File at https://doi.org/10.6084/m9.figshare.23515614) to explain how the interstitial and intrathick ascending limb osmotic concentration (OC) values used in the figures in this article are simulated and how alternative OC values can be generated from Tables S1 and S2 to illustrate CCM.NEW & NOTEWORTHY Countercurrent multiplication is widely accepted as the mechanism for the generation of the corticopapillary osmotic gradient in the outer medulla of mammalian kidneys, but the current explanations of it in textbooks and the literature are inefficient and incomplete, leading to confusion for students. This article shares a modified version of countercurrent multiplication with more understandable explanation as a way of motivating peer discussion, further improvement, and future research.


Asunto(s)
Médula Renal , Riñón , Animales , Humanos , Médula Renal/fisiología , Ósmosis , Mamíferos
2.
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
3.
Biochem Biophys Res Commun ; 514(2): 436-442, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31053298

RESUMEN

Nuclear factor of activated T-cells 5 (NFAT5) directly binds to the promoter of the RING finger protein 183 (RNF183) gene and induces its transcription under hypertonic conditions in mouse inner-medullary collecting duct (mIMCD-3) cells. However, there is no specific anti-RNF183 antibody for immunostaining; therefore, it is unclear whether NFAT5 regulates RNF183 expression in vivo and where RNF183 is localized in the kidney. This study investigated NFAT5-regulated in vivo RNF183 expression and localization using CRISPR/Cas9-mediated RNF183-green fluorescent protein (RNF183-GFP) knock-in mice. GFP with linker sequences was introduced upstream of an RNF183 open reading frame in exon 3 by homologous recombination through a donor plasmid. Immunofluorescence staining using GFP antibody revealed that GFP signals gradually increase from the outer medulla down to the inner medulla and colocalize with aquaporin-2. Furosemide treatment dramatically decreased RNF183 expression in the renal medulla, consistent with the decrease in NFAT5 protein and target gene mRNA expression. Furosemide treatment of mIMCD-3 cells did not affect mRNA expression and RNF183 promoter activities. These results indicated that RNF183 is predominantly expressed in the renal medullary collecting ducts, and that decreased renal medullary tonicity by furosemide treatment decreases RNF183 expression by NFAT5 downregulation.


Asunto(s)
Regulación de la Expresión Génica , Médula Renal/fisiología , Túbulos Renales Colectores/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Regulación hacia Abajo/efectos de los fármacos , Femenino , Furosemida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones
4.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29237738

RESUMEN

Urinary concentrating ability is central to mammalian water balance and depends on a medullary osmotic gradient generated by a countercurrent multiplication mechanism. Medullary hyperosmolarity is protected from washout by countercurrent exchange and efficient removal of interstitial fluid resorbed from the loop of Henle and collecting ducts. In most tissues, lymphatic vessels drain excess interstitial fluid back to the venous circulation. However, the renal medulla is devoid of classic lymphatics. Studies have suggested that the fenestrated ascending vasa recta (AVRs) drain the interstitial fluid in this location, but this function has not been conclusively shown. We report that late gestational deletion of the angiopoietin receptor endothelial tyrosine kinase 2 (Tie2) or both angiopoietin-1 and angiopoietin-2 prevents AVR formation in mice. The absence of AVR associated with rapid accumulation of fluid and cysts in the medullary interstitium, loss of medullary vascular bundles, and decreased urine concentrating ability. In transgenic reporter mice with normal angiopoietin-Tie2 signaling, medullary AVR exhibited an unusual hybrid endothelial phenotype, expressing lymphatic markers (prospero homeobox protein 1 and vascular endothelial growth factor receptor 3) as well as blood endothelial markers (CD34, endomucin, platelet endothelial cell adhesion molecule 1, and plasmalemmal vesicle-associated protein). Taken together, our data redefine the AVRs as Tie2 signaling-dependent specialized hybrid vessels and provide genetic evidence of the critical role of AVR in the countercurrent exchange mechanism and the structural integrity of the renal medulla.


Asunto(s)
Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Líquido Extracelular/metabolismo , Capacidad de Concentración Renal/fisiología , Médula Renal/irrigación sanguínea , Receptor TIE-2/fisiología , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Tipificación del Cuerpo , Linaje de la Célula , Endotelio Vascular , Genes Reporteros , Edad Gestacional , Proteínas de Homeodominio/análisis , Enfermedades Renales Quísticas/genética , Médula Renal/embriología , Médula Renal/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/patología , Ósmosis , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Circulación Renal , Transducción de Señal , Proteínas Supresoras de Tumor/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis
5.
Physiol Genomics ; 50(6): 440-447, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29602296

RESUMEN

Studies exploring the development of hypertension have traditionally been unable to distinguish which of the observed changes are underlying causes from those that are a consequence of elevated blood pressure. In this study, a custom-designed servo-control system was utilized to precisely control renal perfusion pressure to the left kidney continuously during the development of hypertension in Dahl salt-sensitive rats. In this way, we maintained the left kidney at control blood pressure while the right kidney was exposed to hypertensive pressures. As each kidney was exposed to the same circulating factors, differences between them represent changes induced by pressure alone. RNA sequencing analysis identified 1,613 differently expressed genes affected by renal perfusion pressure. Three pathway analysis methods were applied, one a novel approach incorporating arterial pressure as an input variable allowing a more direct connection between the expression of genes and pressure. The statistical analysis proposed several novel pathways by which pressure affects renal physiology. We confirmed the effects of pressure on p-Jnk regulation, in which the hypertensive medullas show increased p-Jnk/Jnk ratios relative to the left (0.79 ± 0.11 vs. 0.53 ± 0.10, P < 0.01, n = 8). We also confirmed pathway predictions of mitochondrial function, in which the respiratory control ratio of hypertensive vs. control mitochondria are significantly reduced (7.9 ± 1.2 vs. 10.4 ± 1.8, P < 0.01, n = 6) and metabolomic profile, in which 14 metabolites differed significantly between hypertensive and control medullas ( P < 0.05, n = 5). These findings demonstrate that subtle differences in the transcriptome can be used to predict functional changes of the kidney as a consequence of pressure elevation.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inflamación/genética , Médula Renal/fisiología , Médula Renal/fisiopatología , Redes y Vías Metabólicas/genética , Perfusión , Animales , Teorema de Bayes , Respiración de la Célula , Hipertensión/genética , Metaboloma , Metabolómica , Mitocondrias/metabolismo , Ratas Endogámicas Dahl , Análisis de Regresión , Programas Informáticos
6.
Am J Physiol Renal Physiol ; 309(7): F627-37, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26290371

RESUMEN

The architecture of the inner stripe of the outer medulla of the human kidney has long been known to exhibit distinctive configurations; however, inner medullary architecture remains poorly defined. Using immunohistochemistry with segment-specific antibodies for membrane fluid and solute transporters and other proteins, we identified a number of distinctive functional features of human inner medulla. In the outer inner medulla, aquaporin-1 (AQP1)-positive long-loop descending thin limbs (DTLs) lie alongside descending and ascending vasa recta (DVR, AVR) within vascular bundles. These vascular bundles are continuations of outer medullary vascular bundles. Bundles containing DTLs and vasa recta lie at the margins of coalescing collecting duct (CD) clusters, thereby forming two regions, the vascular bundle region and the CD cluster region. Although AQP1 and urea transporter UT-B are abundantly expressed in long-loop DTLs and DVR, respectively, their expression declines with depth below the outer medulla. Transcellular water and urea fluxes likely decline in these segments at progressively deeper levels. Smooth muscle myosin heavy chain protein is also expressed in DVR of the inner stripe and the upper inner medulla, but is sparsely expressed at deeper inner medullary levels. In rodent inner medulla, fenestrated capillaries abut CDs along their entire length, paralleling ascending thin limbs (ATLs), forming distinct compartments (interstitial nodal spaces; INSs); however, in humans this architecture rarely occurs. Thus INSs are relatively infrequent in the human inner medulla, unlike in the rodent where they are abundant. UT-B is expressed within the papillary epithelium of the lower inner medulla, indicating a transcellular pathway for urea across this epithelium.


Asunto(s)
Médula Renal/anatomía & histología , Médula Renal/fisiología , Acuaporina 1/metabolismo , Capilares/metabolismo , Epitelio/metabolismo , Humanos , Imagenología Tridimensional , Inmunohistoquímica , Técnicas In Vitro , Capacidad de Concentración Renal/fisiología , Túbulos Renales/metabolismo , Túbulos Renales Colectores/metabolismo , Consumo de Oxígeno
7.
Kidney Int ; 88(5): 1079-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26108066

RESUMEN

Chronic activation of the renin-angiotensin system promotes hypertension, renal microvascular dysfunction, tissue hypoxia, and inflammation. Despite similar hypertension, an injurious response to excess angiotensin II is greater in F344 than in Lewis rats; the latter displaying renoprotection. Here we studied whether p2rx7, encoding the P2X7 receptor (P2X7R), is a candidate gene for the differential susceptibility to vascular dysfunction under high angiotensin II tone. A 14-day infusion of angiotensin II into F344 rats increased blood pressure by about 15 mm Hg without inducing fibrosis or albuminuria. In vivo pressure natriuresis was suppressed, medullary perfusion reduced by half, and the corticomedullary oxygenation gradient disrupted. Selective P2X7R antagonism restored pressure natriuresis, promoting a significant leftward shift in the intercept and increasing the slope. Sodium excretion was increased sixfold and blood pressure normalized. The specific P2X7R antagonist AZ11657312 increased renal medullary perfusion, but only in angiotensin II-treated rats. Tissue oxygenation was improved by P2X7R blockade, particularly in poorly oxygenated regions of the kidney. Thus, activation of P2X7R induces microvascular dysfunction and regional hypoxia when angiotensin II is elevated and these effects may contribute to progression of renal injury induced by chronic angiotensin II.


Asunto(s)
Corteza Renal/irrigación sanguínea , Médula Renal/irrigación sanguínea , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/metabolismo , Circulación Renal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/metabolismo , Expresión Génica/efectos de los fármacos , Corteza Renal/fisiología , Médula Renal/fisiología , Masculino , Natriuresis/efectos de los fármacos , Óxido Nítrico/metabolismo , Oxígeno/sangre , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores Purinérgicos P2X7/genética , Vasoconstrictores/farmacología
8.
J Magn Reson Imaging ; 40(5): 1099-102, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24925441

RESUMEN

PURPOSE: To assess the feasibility of diffusion tensor imaging (DTI) of normal kidneys and the influence of hydration state. MATERIALS AND METHODS: Ten healthy volunteers underwent renal DTI after fasting for 12 hours and 4 hours, without fasting, and following water diuresis. Medullary and cortical apparent diffusion coefficient (ADC) and fractional anisotropy (FA) values were measured and compared in the four different states of hydration. DTI was performed with a 3T magnetic resonance imaging (MRI) system using fat-saturated single-shot spin-echo echo planar imaging sequence. RESULTS: ADC of normal cortex (2.387 ± 0.081 × 10(-3) mm(2) /s) was significantly higher (t = 20.126, P = 0) than that of medulla (1.990 ± 0.063 × 10(-3) mm(2) /s). The FA value of normal cortex (0.282 ± 0.017) was significantly lower (t = -42.713, P = 0) than that of medulla (0.447 ± 0.022). The ADC and FA values of the left renal cortex (2.404 ± 0.082 × 10(-3) mm(2) /s, 0.282 ± 0.017) and medulla (2.002 ± 0.081 × 10(-3) mm(2) /s, 0.452 ± 0.024) were not significantly different (P > 0.05) from those of right renal cortex (2.369 ± 0.080 × 10(-3) mm(2) /s, 0.283 ± 0.018) and medulla (1.978 ± 0.039 × 10(-3) mm(2) /s, 0.443 ± 0.019). Values for ADC (×10(-3) mm(2) /s) and FA in the 12-hour fasting, 4-hour fasting, nonfasting, and water diuresis states were 2.372 ± 0.095 and 0.278 ± 0.018, 2.387 ± 0.081 and 0.282 ± 0.017, 2.416 ± 0.051 and 0.279 ± 0.023, 2.421 ± 0.068, and 0.270 ± 0.021, respectively, in cortex, 1.972 ± 0.084 and 0.438 ± 0.014, 1.990 ± 0.063 and 0.447 ± 0.022, 2.021 ± 0.081 and 0.450 ± 0.031, 2.016 ± 0.076 and 0.449 ± 0.028, respectively, in medulla. The ADC and FA values in different hydration states were not significantly different (P > 0.05). CONCLUSION: DTI of normal kidneys is feasible with reproducible ADC and FA values independent of hydration states.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Imagen Eco-Planar/métodos , Aumento de la Imagen/métodos , Corteza Renal/anatomía & histología , Corteza Renal/fisiología , Médula Renal/anatomía & histología , Médula Renal/fisiología , Adulto , Anisotropía , Agua Corporal/fisiología , Diuresis/fisiología , Ayuno , Femenino , Humanos , Masculino , Valores de Referencia
9.
Am J Physiol Renal Physiol ; 305(9): F1298-305, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23986514

RESUMEN

The Gi-coupled adenine receptor (AdeR) binds adenine with high affinity and potentially reduces cellular cAMP levels. Since cAMP is an important second messenger in the renal transport of water and solutes, we localized AdeR in the rat kidney. Real-time RT-PCR showed higher relative expression of AdeR mRNA in the cortex and outer medulla compared with the inner medulla. Immunoblots using a peptide-derived and affinity-purified rabbit polyclonal antibody specific for an 18-amino acid COOH-terminal sequence of rat AdeR, which we generated, detected two bands between ∼30 and 40 kDa (molecular mass of native protein: 37 kDa) in the cortex, outer medulla, and inner medulla. These bands were ablated by preadsorption of the antibody with the immunizing peptide. Immunofluorescence labeling showed expression of AdeR protein in all regions of the kidney. Immunoperoxidase revealed strong labeling of AdeR protein in the cortical vasculature, including the glomerular arterioles, and less intense labeling in the cells of the collecting duct system. Confocal immunofluorescence imaging colocalized AdeR with aquaporin-2 protein to the apical plasma membrane in the collecting duct. Functionally, adenine (10 µM) significantly decreased (P < 0.01) 1-deamino-8-d-arginine vasopressin (10 nM)-induced cAMP production in ex vivo preparations of inner medullary collecting ducts, which was reversed by PSB-08162 (20 µM, P < 0.01), a selective antagonist of AdeR. Thus, we demonstrated the expression of AdeR in the renal vasculature and collecting ducts and its functional relevance. This study may open a new avenue for the exploration of autocrine/paracrine regulation of renal vascular and tubular functions by the nucleobase adenine in health and disease.


Asunto(s)
Médula Renal/metabolismo , Túbulos Renales Colectores/metabolismo , Receptores Purinérgicos/metabolismo , Animales , AMP Cíclico/metabolismo , Desamino Arginina Vasopresina/metabolismo , Médula Renal/fisiología , Túbulos Renales Colectores/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos/fisiología
10.
Am J Physiol Renal Physiol ; 304(2): F233-8, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23097465

RESUMEN

The kidney's ability to concentrate urine is vitally important to our quality of life. In the hypertonic environment of the kidney, urea transporters must be regulated to optimize function. We previously showed that hypertonicity increases urea permeability and that the protein kinase C (PKC) blockers chelerythrine and rottlerin decreased hypertonicity-stimulated urea permeability in rat inner medullary collecting ducts (IMCDs). Because PKCα knockout (PKCα(-/-)) mice have a urine-concentrating defect, we tested the effect of hypertonicity on urea permeability in isolated perfused mouse IMCDs. Increasing the osmolality of perfusate and bath from 290 to 690 mosmol/kgH(2)O did not change urea permeability in PKCα(-/-) mice but significantly increased urea permeability in wild-type mice. To determine whether the response to protein kinase A was also missing in IMCDs of PKCα(-/-) mice, tubules were treated with vasopressin and subsequently with the PKC stimulator phorbol dibutyrate (PDBu). Vasopressin stimulated urea permeability in PKCα(-/-) mice. Like vasopressin, forskolin stimulated urea permeability in PKCα(-/-) mice. We previously showed that, in rats, vasopressin and PDBu have additive stimulatory effects on urea permeability. In contrast, in PKCα(-/-) mice, PDBu did not further increase vasopressin-stimulated urea permeability. Western blot analysis showed that expression of the UT-A1 urea transporter in IMCDs was increased in response to vasopressin in wild-type mice as well as PKCα(-/-) mice. Hypertonicity increased UT-A1 phosphorylation in wild-type mice but not in PKCα(-/-) mice. We conclude that PKCα mediates hypertonicity-stimulated urea transport but is not necessary for vasopressin stimulation of urea permeability in mouse IMCDs.


Asunto(s)
Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/metabolismo , Proteína Quinasa C-alfa/metabolismo , Solución Salina Hipertónica/farmacología , Urea/metabolismo , Animales , Transporte Biológico , Colforsina/farmacología , Médula Renal/anatomía & histología , Médula Renal/fisiología , Ratones , Ratones Noqueados , Permeabilidad , Forbol 12,13-Dibutirato , Proteína Quinasa C-alfa/genética , Vasopresinas/metabolismo , Vasopresinas/farmacología
11.
Am J Physiol Regul Integr Comp Physiol ; 304(7): R488-503, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364530

RESUMEN

Comparative studies of renal structure and function have potential to provide insights into the urine-concentrating mechanism of the mammalian kidney. This review focuses on the tubular transport pathways for water and urea that play key roles in fluid and solute movements between various compartments of the rodent renal inner medulla. Information on aquaporin water channel and urea transporter expression has increased our understanding of functional segmentation of medullary thin limbs of Henle's loops, collecting ducts, and vasa recta. A more complete understanding of membrane transporters and medullary architecture has identified new and potentially significant interactions between these structures and the interstitium. These interactions are now being introduced into our concept of how the inner medullary urine-concentrating mechanism works. A variety of regulatory pathways lead directly or indirectly to variable patterns of fluid and solute movements among the interstitial and tissue compartments. Animals with the ability to produce highly concentrated urine, such as desert species, are considered to exemplify tubular structure and function that optimize urine concentration. These species may provide unique insights into the urine-concentrating process.(1)


Asunto(s)
Médula Renal/fisiología , Roedores/anatomía & histología , Roedores/fisiología , Urea/metabolismo , Agua/metabolismo , Animales , Transporte Biológico/fisiología , Médula Renal/anatomía & histología , Médula Renal/irrigación sanguínea , Nefronas/anatomía & histología , Nefronas/irrigación sanguínea , Nefronas/fisiología
12.
Biochim Biophys Acta ; 1814(3): 435-48, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21236367

RESUMEN

The papilla of the mammalian kidney must tolerate greatly varying degrees of hyperosmotic stress during urine concentration and depending on whole organism hydration state. To identify proteome adaptations supporting cell function and survival in such a harsh environment we compared the proteome of a) the hyperosmotic renal papilla with that of adjacent iso-osmotic cortex tissue and b) the renal papilla of diuretic versus that of anti-diuretic mice. Though functionally distinct the papilla is in close physical proximity to the renal cortex, an iso-osmotic region. Proteomic differences between the papilla and cortex of C57BL6 mice were identified using two-dimensional gel electrophoresis and MALDI-TOF/TOF mass spectrometry. We found 37 different proteins characteristic of the cortex and 16 proteins over-represented in the papilla. Regional specificity was confirmed by Western blot and further substantiated by immunohistochemistry for selected proteins. Proteins that are characteristic of the renal papilla include αB crystallin, Hsp beta-1, Hsp90, 14-3-3 protein, glutathione S-transferase, aldose reductase, actin and tropomyosin. Gene ontology analysis confirmed a significant increase in molecular functions associated with protein chaperoning and cell stabilization. Proteins over-represented in the cortex were largely related to routine metabolism. During antidiuresis 15 different proteins changed significantly while 18 different proteins changed significantly during diuresis relative to normally hydrated controls. Changes were confirmed by Western blot for selected proteins. Proteins that are significantly altered by diuretic state are associated with cell structure (actin, tubulin), signaling (Rho GDP dissociation inhibitor, abhydrolase domain-containing protein 14B), chaperone functioning (Hsp beta-1, αB crystallin, T complex protein-1) and anti-oxidant functions (α-enolase, GAPDH and LDH). Taken together our study reveals that specific proteins involved in protein folding, cytoskeletal stabilization, antioxidant responses, and stress signaling contribute greatly to the unique hyperosmotic stress resistant phenotype of the kidney papilla.


Asunto(s)
Corteza Renal/fisiología , Médula Renal/fisiología , Ósmosis/efectos de los fármacos , Animales , Desamino Arginina Vasopresina/farmacología , Diuresis/efectos de los fármacos , Diuresis/fisiología , Femenino , Perfilación de la Expresión Génica , Médula Renal/metabolismo , Masculino , Ratones , Concentración Osmolar , Proteoma/metabolismo , Proteómica/métodos
13.
Am J Physiol Renal Physiol ; 302(7): F830-9, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22160770

RESUMEN

Recent anatomic findings indicate that in the upper inner medulla of the rodent kidney, tubules, and vessels are organized around clusters of collecting ducts (CDs). Within CD clusters, CDs and some of the ascending vasa recta (AVR) and ascending thin limbs (ATLs), when viewed in transverse sections, form interstitial nodal spaces, which are arrayed at structured intervals throughout the inner medulla. These spaces, or microdomains, are bordered on one side by a single CD, on the opposite side by one or more ATLs, and on the other two sides by AVR. To study the interactions among these CDs, ATLs, and AVR, we have developed a mathematical compartment model, which simulates steady-state solute exchange through the microdomain at a given inner medullary level. Fluid in all compartments contains Na(+), Cl(-), urea and, in the microdomain, negative fixed charges that represent macromolecules (e.g., hyaluronan) balanced by Na(+). Fluid entry into AVR is assumed to be driven by hydraulic and oncotic pressures. Model results suggest that the isolated microdomains facilitate solute and fluid mixing among the CDs, ATLs, and AVR, promote water withdrawal from CDs, and consequently may play an important role in generating the inner medullary osmotic gradient.


Asunto(s)
Médula Renal/fisiología , Túbulos Renales Colectores/fisiología , Asa de la Nefrona/fisiología , Modelos Biológicos , Animales , Permeabilidad Capilar , Simulación por Computador , Ácido Hialurónico/metabolismo , Presión Hidrostática , Inmunohistoquímica , Asa de la Nefrona/irrigación sanguínea , Masculino , Ratas , Ratas Wistar , Sodio/metabolismo , Urea/metabolismo , Agua/metabolismo
14.
Am J Physiol Renal Physiol ; 302(3): F316-28, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22071246

RESUMEN

The Na(+)- and Cl(-)-dependent GABA-betaine transporter (BGT1) has received attention mostly as a protector against osmolarity changes in the kidney and as a potential controller of the neurotransmitter GABA in the brain. Nevertheless, the cellular distribution of BGT1, and its physiological importance, is not fully understood. Here we have quantified mRNA levels using TaqMan real-time PCR, produced a number of BGT1 antibodies, and used these to study BGT1 distribution in mice. BGT1 (protein and mRNA) is predominantly expressed in the liver (sinusoidal hepatocyte plasma membranes) and not in the endothelium. BGT1 is also present in the renal medulla, where it localizes to the basolateral membranes of collecting ducts (particularly at the papilla tip) and the thick ascending limbs of Henle. There is some BGT1 in the leptomeninges, but brain parenchyma, brain blood vessels, ependymal cells, the renal cortex, and the intestine are virtually BGT1 deficient in 1- to 3-mo-old mice. Labeling specificity was assured by processing tissue from BGT1-deficient littermates in parallel as negative controls. Addition of 2.5% sodium chloride to the drinking water for 48 h induced a two- to threefold upregulation of BGT1, tonicity-responsive enhancer binding protein, and sodium-myo-inositol cotransporter 1 (slc5a3) in the renal medulla, but not in the brain and barely in the liver. BGT1-deficient and wild-type mice appeared to tolerate the salt treatment equally well, possibly because betaine is one of several osmolytes. In conclusion, this study suggests that BGT1 plays its main role in the liver, thereby complementing other betaine-transporting carrier proteins (e.g., slc6a20) that are predominantly expressed in the small intestine or kidney rather than the liver.


Asunto(s)
Encéfalo/fisiología , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Riñón/fisiología , Hígado/fisiología , Animales , Anticuerpos/farmacología , Membrana Celular/fisiología , Proteínas Transportadoras de GABA en la Membrana Plasmática/inmunología , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Células HEK293 , Hepatocitos/fisiología , Humanos , Médula Renal/fisiología , Túbulos Renales Colectores/fisiología , Hígado/citología , Asa de la Nefrona/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Presión Osmótica/fisiología , ARN Mensajero/metabolismo , Conejos , Cloruro de Sodio/farmacología
15.
Am J Physiol Regul Integr Comp Physiol ; 303(7): R748-56, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22914749

RESUMEN

We hypothesize that the inner medulla of the kangaroo rat Dipodomys merriami, a desert rodent that concentrates its urine to over 6,000 mosmol/kg H(2)O, provides unique examples of architectural features necessary for production of highly concentrated urine. To investigate this architecture, inner medullary vascular segments in the outer inner medulla were assessed with immunofluorescence and digital reconstructions from tissue sections. Descending vasa recta (DVR) expressing the urea transporter UT-B and the water channel aquaporin 1 lie at the periphery of groups of collecting ducts (CDs) that coalesce in their descent through the inner medulla. Ascending vasa recta (AVR) lie inside and outside groups of CDs. DVR peel away from vascular bundles at a uniform rate as they descend the inner medulla, and feed into networks of AVR that are associated with organized clusters of CDs. These AVR form interstitial nodal spaces, with each space composed of a single CD, two AVR, and one or more ascending thin limbs or prebend segments, an architecture that may lead to solute compartmentation and fluid fluxes essential to the urine concentrating mechanism. Although we have identified several apparent differences, the tubulovascular architecture of the kangaroo rat inner medulla is remarkably similar to that of the Munich Wistar rat at the level of our analyses. More detailed studies are required for identifying interspecies functional differences.


Asunto(s)
Dipodomys/anatomía & histología , Dipodomys/fisiología , Capacidad de Concentración Renal/fisiología , Médula Renal/anatomía & histología , Médula Renal/irrigación sanguínea , Animales , Acuaporina 1/metabolismo , Capilares/anatomía & histología , Capilares/citología , Capilares/fisiología , Femenino , Médula Renal/fisiología , Túbulos Renales Colectores/anatomía & histología , Túbulos Renales Colectores/irrigación sanguínea , Túbulos Renales Colectores/metabolismo , Masculino , Proteínas de Transporte de Membrana/metabolismo , Modelos Animales , Ratas , Ratas Wistar , Flujo Sanguíneo Regional/fisiología , Vasopresinas/sangre , Transportadores de Urea
16.
Nephron Physiol ; 122(1-2): 7-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23548701

RESUMEN

BACKGROUND/AIMS: Since furosemide (FS) inhibits active Na(+) reabsorption by medullary thick ascending limb (mTAL) in the renal outer medulla, it may decrease its work during periods of low O2 supply to deep in the renal outer medulla. This study was designed to demonstrate that there may be a dose of FS would reduce its metabolic work while preventing the excessive loss of magnesium (Mg(2+)). Mg(2+) is important because the ATP needed to perform work must have bound Mg(2+) to it. METHODS: Rats were injected intraperitoneally with a range of doses of FS. The measured outcomes were urine flow rate and parameters of functions of the mTAL (i.e. urine and renal papillary osmolality and urinary excretion of Na(+), Cl(-), K(+) and Mg(2+), and concentrations of Mg(2+) in serum). RESULTS: The urine flow rate increased significantly starting at 2.4 mg FS/kg. The renal papillary osmolality decreased at ≥0.4 mg FS/kg, and the large detectable natriuresis started at 1.6 mg FS/kg. At this latter dose, the urinary excretion of Mg(2+) rose significantly. CONCLUSION: In rats, the non-natriuretic dose of FS may reduce the work of the mTAL. The earliest indicator of reduced work in the mTAL appears to be a decrease in urine osmolality rather than a rise in urine flow rate. Higher doses of FS should be avoided, as they induce high rates of Mg(2+) excretion, which can deplete the body of this essential electrolyte.


Asunto(s)
Furosemida/farmacología , Médula Renal/efectos de los fármacos , Magnesio/orina , Animales , Cloruros/orina , Diuréticos/administración & dosificación , Diuréticos/farmacología , Relación Dosis-Respuesta a Droga , Furosemida/administración & dosificación , Inyecciones Intraperitoneales , Médula Renal/fisiología , Magnesio/sangre , Masculino , Natriuresis/efectos de los fármacos , Concentración Osmolar , Potasio/orina , Ratas , Ratas Sprague-Dawley , Sodio/orina , Orina/química , Urodinámica/efectos de los fármacos
17.
Am J Physiol Renal Physiol ; 300(2): F356-71, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21068086

RESUMEN

A new, region-based mathematical model of the urine concentrating mechanism of the rat renal medulla was used to investigate the significance of transport and structural properties revealed in anatomic studies. The model simulates preferential interactions among tubules and vessels by representing concentric regions that are centered on a vascular bundle in the outer medulla (OM) and on a collecting duct cluster in the inner medulla (IM). Particularly noteworthy features of this model include highly urea-permeable and water-impermeable segments of the long descending limbs and highly urea-permeable ascending thin limbs. Indeed, this is the first detailed mathematical model of the rat urine concentrating mechanism that represents high long-loop urea permeabilities and that produces a substantial axial osmolality gradient in the IM. That axial osmolality gradient is attributable to the increasing urea concentration gradient. The model equations, which are based on conservation of solutes and water and on standard expressions for transmural transport, were solved to steady state. Model simulations predict that the interstitial NaCl and urea concentrations in adjoining regions differ substantially in the OM but not in the IM. In the OM, active NaCl transport from thick ascending limbs, at rates inferred from the physiological literature, resulted in a concentrating effect such that the intratubular fluid osmolality of the collecting duct increases ~2.5 times along the OM. As a result of the separation of urea from NaCl and the subsequent mixing of that urea and NaCl in the interstitium and vasculature of the IM, collecting duct fluid osmolality further increases by a factor of ~1.55 along the IM.


Asunto(s)
Capacidad de Concentración Renal/fisiología , Médula Renal/fisiología , Modelos Biológicos , Animales , Transporte Biológico Activo/fisiología , Simulación por Computador , Túbulos Renales/fisiología , Asa de la Nefrona/fisiología , Concentración Osmolar , Ratas , Cloruro de Sodio/metabolismo , Urea/metabolismo , Agua/metabolismo
18.
Am J Physiol Renal Physiol ; 300(2): F372-84, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21068088

RESUMEN

In a companion study [Layton AT. A mathematical model of the urine concentrating mechanism in the rat renal medulla. I. Formulation and base-case results. Am J Physiol Renal Physiol. (First published November 10, 2010). 10.1152/ajprenal.00203.2010] a region-based mathematical model was formulated for the urine concentrating mechanism in the renal medulla of the rat kidney. In the present study, we investigated model sensitivity to some of the fundamental structural assumptions. An unexpected finding is that the concentrating capability of this region-based model falls short of the capability of models that have radially homogeneous interstitial fluid at each level of only the inner medulla (IM) or of both the outer medulla and IM, but are otherwise analogous to the region-based model. Nonetheless, model results reveal the functional significance of several aspects of tubular segmentation and heterogeneity: 1) the exclusion of ascending thin limbs that reach into the deep IM from the collecting duct clusters in the upper IM promotes urea cycling within the IM; 2) the high urea permeability of the lower IM thin limb segments allows their tubular fluid urea content to equilibrate with the surrounding interstitium; 3) the aquaporin-1-null terminal descending limb segments prevent water entry and maintain the transepithelial NaCl concentration gradient; 4) a higher thick ascending limb Na(+) active transport rate in the inner stripe augments concentrating capability without a corresponding increase in energy expenditure for transport; 5) active Na(+) reabsorption from the collecting duct elevates its tubular fluid urea concentration. Model calculations predict that these aspects of tubular segmentation and heterogeneity promote effective urine concentrating functions.


Asunto(s)
Capacidad de Concentración Renal/fisiología , Médula Renal/anatomía & histología , Médula Renal/fisiología , Modelos Biológicos , Animales , Acuaporina 1/análisis , Acuaporina 1/metabolismo , Transporte Biológico Activo/fisiología , Simulación por Computador , Asa de la Nefrona/fisiología , Concentración Osmolar , Ratas , Cloruro de Sodio/metabolismo , Urea/metabolismo
19.
Am J Physiol Renal Physiol ; 301(1): F35-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478479

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite formed by phosphorylation of sphingosine. S1P has been indicated to play a significant role in the cardiovascular system. It has been shown that the enzymes for S1P metabolism are expressed in the kidneys. The present study characterized the expression of S1P receptors in the kidneys and determined the role of S1P in the control of renal hemodynamics and sodium excretion. Real-time RT-PCR analyses showed that S1P receptors S1P1, S1P2, and S1P3 were most abundantly expressed in the renal medulla. Immunohistochemistry revealed that all three types of S1P receptors were mainly located in collecting ducts. Intramedullary infusion of FTY720, an S1P agonist, produced a dramatic increase in sodium excretion by twofold and a small but significant increase in medullary blood flow (16%). Administration of W146, an S1P1 antagonist, into the renal medulla blocked the effect of FTY720 and decreased the sodium excretion by 37% when infused alone. The antagonists of S1P2 and S1P3 had no effect. FTY720 produced additive natriuretic effects in combination with different sodium transporter inhibitors except amiloride, an epithelial sodium channel blocker. In the presence of nitric oxide synthase inhibitor l-NAME, FTY720 still increased sodium excretion. These data suggest that S1P produces natriuretic effects via activation of S1P1 in the renal medulla and this natriuretic effect may be through inhibition of epithelial sodium channel, which is nitric oxide independent. It is concluded that S1P is a novel diuretic factor in the renal medulla and may be an important regulator of sodium homeostasis.


Asunto(s)
Médula Renal/fisiología , Lípidos/fisiología , Lisofosfolípidos/fisiología , Natriuréticos/fisiología , Esfingosina/análogos & derivados , Animales , Inhibidores Enzimáticos/farmacología , Clorhidrato de Fingolimod , Inmunohistoquímica , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Médula Renal/irrigación sanguínea , Masculino , NG-Nitroarginina Metil Éster/farmacología , Glicoles de Propileno/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología , Circulación Renal/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/orina , Esfingosina/farmacología , Esfingosina/fisiología
20.
Clin Exp Pharmacol Physiol ; 38(4): 247-54, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21306412

RESUMEN

1. Oxygen tension (PO(2)) in renal cortical tissue can remain relatively constant when renal blood flow changes in the physiological range, even when changes in renal oxygen delivery (DO(2)) and oxygen consumption (VO(2)) are mismatched. In the current study, we examined whether this also occurs in the renal medulla and skeletal muscle, or if it is an unusual property of the renal cortex. We also examined the potential for dysfunction of the mechanisms underlying this phenomenon to contribute to kidney hypoxia in disease states associated with increased renal VO(2) . 2. In both the kidney and hindlimb of pentobarbitone anaesthetized rabbits, whole organ blood flow was reduced by intra-arterial infusion of angiotensin-II and increased by acetylcholine infusion. In the kidney, this was carried out before and during renal arterial infusion of the mitochondrial uncoupler, 2,4-dinitrophenol (DNP), or its vehicle. 3. Angiotensin-II reduced renal (-34%) and hindlimb (-25%) DO(2) , whereas acetylcholine increased renal (+38%) and hindlimb (+66%) DO(2) . However, neither renal nor hindlimb VO(2) were altered. Tissue PO(2) varied with local perfusion in the renal medulla and biceps femoris, but not the renal cortex. DNP increased renal VO(2) (+38%) and reduced cortical tissue PO(2) (-44%), but both still remained stable during subsequent infusion of angiotensin-II and acetylcholine. 4. We conclude that maintenance of tissue PO(2) in the face of mismatched changes in local perfusion and VO(2) is an unusual property of the renal cortex. The underlying mechanisms remain unknown, but our current findings suggest they are not compromised when resting renal VO(2) is increased.


Asunto(s)
Corteza Renal/irrigación sanguínea , Consumo de Oxígeno/fisiología , Oxígeno/sangre , 2,4-Dinitrofenol/farmacología , Acetilcolina/farmacología , Angiotensina II/farmacología , Animales , Hipoxia de la Célula/fisiología , Miembro Posterior/irrigación sanguínea , Miembro Posterior/efectos de los fármacos , Infusiones Intraarteriales , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Médula Renal/irrigación sanguínea , Médula Renal/efectos de los fármacos , Médula Renal/fisiología , Masculino , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/fisiología , Presión Parcial , Perfusión , Conejos , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA