Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 726
Filtrar
1.
Adv Physiol Educ ; 47(3): 665-671, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37439318

RESUMO

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.


Assuntos
Medula Renal , Rim , Animais , Humanos , Medula Renal/fisiologia , Osmose , Mamíferos
2.
Biol Sex Differ ; 11(1): 52, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32928299

RESUMO

BACKGROUND: Premenopausal women have a lower risk of hypertension compared to age-matched men and postmenopausal women. P2Y2 and P2Y4 purinoceptor can be considered potential contributors to hypertension due to their emerging roles in regulating renal tubular Na+ transport. Activation of these receptors inhibits epithelial Na+ channel activity (ENaC) via a phospholipase C (PLC)-dependent pathway resulting in natriuresis. We recently reported that activation of P2Y2 and P2Y4 receptors in the renal medulla by UTP promotes natriuresis in male and ovariectomized (OVX) rats, but not in ovary-intact females. This led us to hypothesize that ovary-intact females have greater basal renal medullary activity of P2 (P2Y2 and P2Y4) receptors regulating Na+ excretion compared to male and OVX rats. METHODS: To test our hypothesis, we determined (i) the effect of inhibiting medullary P2 receptors by suramin (750 µg/kg/min) on urinary Na+ excretion in anesthetized male, ovary-intact female, and OVX Sprague Dawley rats, (ii) mRNA expression and protein abundance of P2Y2 and P2Y4 receptors, and (iii) mRNA expression of their downstream effectors (PLC-1δ and ENaCα) in renal inner medullary tissues obtained from these three groups. We also subjected cultured mouse inner medullary collecting duct cells (segment 3, mIMCD3) to different concentrations of 17ß-estradiol (E2, 0, 10, 100, and 1000 nM) to test whether E2 increases mRNA expression of P2Y2 and P2Y4 receptors. RESULTS: Acute P2 inhibition attenuated urinary Na+ excretion in ovary-intact females, but not in male or OVX rats. We found that P2Y2 and P2Y4 mRNA expression was higher in the inner medulla from females compared to males or OVX. Inner medullary lysates showed that ovary-intact females have higher P2Y2 receptor protein abundance, compared to males; however, OVX did not eliminate this sex difference. We also found that E2 dose-dependently upregulated P2Y2 and P2Y4 mRNA expression in mIMCD3. CONCLUSION: These data suggest that ovary-intact females have enhanced P2Y2 and P2Y4-dependent regulation of Na+ handling in the renal medulla, compared to male and OVX rats. We speculate that the P2 pathway contributes to facilitated renal Na+ handling in premenopausal females.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Estradiol/metabolismo , Natriurese/fisiologia , Ovário/fisiologia , Receptores Purinérgicos P2Y2/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Canais Epiteliais de Sódio/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Medula Renal/fisiologia , Masculino , Ovariectomia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y2/genética , Fatores Sexuais , Suramina/farmacologia , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo
3.
J Am Soc Nephrol ; 31(7): 1555-1568, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32487560

RESUMO

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.


Assuntos
Pressão Sanguínea , Medula Renal/fisiologia , Receptor de Endotelina A/fisiologia , Aldosterona/sangue , Animais , Arginina Vasopressina/urina , Cálcio/metabolismo , Diurese/efeitos dos fármacos , Endotelina-1/farmacologia , Endotelina-1/urina , Canais Epiteliais de Sódio/metabolismo , Feminino , Genótipo , Taxa de Filtração Glomerular , Ácido Hialurônico/metabolismo , Medula Renal/citologia , Medula Renal/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Natriurese/efeitos dos fármacos , Nitratos/urina , Nitritos/urina , Potássio/urina , RNA Mensageiro/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Sódio/urina , Cloreto de Sódio na Dieta/administração & dosagem , Tamoxifeno/farmacologia , Água/administração & dosagem , Água/metabolismo
4.
PLoS One ; 14(11): e0225640, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31774858

RESUMO

We recently provided highly suggestive preliminary evidence that the renal interstitium contracts reactively in vivo. We demonstrated that renal medullary direct interstitial volume expansion (rmDIVE = 100 µl bolus infusion of 0.9% saline (SS)/30 s) brought about a biphasic renal interstitial hydrostatic pressure (RIHP) response which was abolished when dibutyryl-cAMP was concomitant and interstitially infused. To assess more deeply the feasibility of the concept that the renal interstitium contracts in vivo, two experimental series (S1, S2) were performed in hydropenic rats subjected to acute left renal-denervation, hormonal clamping, and control of renal arterial pressure. In S1, RIHP and renal outer medullary blood flow (RoMBF) were continuously measured before and after a sudden micro-bolus (5µl) injection, into the renal medullary interstitium, of SS containing α-trinositol (α-TNS, anti-inflammatory drug) to either two doses 2 or 4 mM (SS + 2 α-TNS and SS + 4 α-TNS groups). No overall differences between groups in either ΔRIHP or %ΔRoMBF time courses were found; however, in the SS + 2 α-TNS group the data were less scattered and the ΔRIHP time course tended to peak faster and then persisted there, so that, this α-TNS dose was selected for S2. In S2, RIHP and RoMBF were similarly measured in rats randomly assigned to three groups: the CTR group (sham time-control), SS group (SS alone), and SS + α-TNS group. The micro-bolus injection of SS alone (SS group) was unable to increase ΔRIHP. The group with no micro-bolus injection (CTR group) experienced a decrease in ΔRIHP. The micro-bolus injection of SS + 2 α-TNS was accompanied by a differential increase in ΔRIHP (vs. CTR and SS groups). These responses were not associated with differential changes among groups in %ΔRoMBF or hemodilution parameters. These results provide additional evidence that the renal interstitium contracts in vivo.


Assuntos
Medula Renal/fisiologia , Circulação Renal , Vasoconstrição/fisiologia , Animais , Pressão Hidrostática , Medula Renal/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Cloreto de Sódio/administração & dosagem , Vasoconstrição/efeitos dos fármacos
5.
Biochem Biophys Res Commun ; 514(2): 436-442, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31053298

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Medula Renal/fisiologia , Túbulos Renais Coletores/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Regulação para Baixo/efeitos dos fármacos , Feminino , Furosemida/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos
6.
Acta Physiol (Oxf) ; 227(1): e13292, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31046189

RESUMO

AIM: Disturbances of renal medullary perfusion and metabolism have been implicated in the pathogenesis of kidney disease and hypertension. Furosemide, a loop diuretic, is widely used to prevent renal medullary hypoxia in acute kidney disease by uncoupling sodium metabolism, but its effects on medullary perfusion in humans are unknown. We performed quantitative imaging of both renal perfusion and oxygenation using Magnetic Resonance Imaging (MRI) before and during furosemide. Based on the literature, we hypothesized that furosemide would increase medullary oxygenation, decrease medullary perfusion, but cause minor changes (<10%) in renal artery flow (RAF). METHODS: Interleaved measurements of RAF, oxygenation (T2 *) and perfusion by arterial spin labelling in the renal cortex and medulla of 9 healthy subjects were acquired before and after an injection of 20 mg furosemide. They were preceded by measurements made during isometric exercise (5 minutes handgrip bouts), which are known to induce changes in renal hemodynamics, that served as a control for the sensitivity of the hemodynamic MRI measurements. Experiments were repeated on a second day to establish that the measurements and the induced changes were reproducible. RESULTS: After furosemide, T2 * values in the medulla increased by 53% (P < 0.01) while RAF and perfusion remained constant. After hand-grip exercise, T2 * values in renal medulla increased by 22% ± 9% despite a drop in medullary perfusion of 7.2% ± 4.7% and a decrease in renal arterial flow of 17.5% ± 1.7% (P < 0.05). Mean coefficients of variation between repeated measurements for all parameters were 7%. CONCLUSION: Furosemide induced the anticipated increase in renal medullary oxygenation, attributable exclusively to a decrease in renal oxygen consumption, since no change of RAF, cortical or medullary perfusion could be demonstrated. All measures and the induced changes were reproducible.


Assuntos
Diuréticos/farmacologia , Furosemida/farmacologia , Córtex Renal/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Adulto , Feminino , Humanos , Córtex Renal/fisiologia , Medula Renal/fisiologia , Masculino , Pessoa de Meia-Idade , Consumo de Oxigênio/fisiologia , Adulto Jovem
7.
Sci Rep ; 9(1): 4692, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30886169

RESUMO

Because exosomes have gained attention as a source of biomarkers, we investigated if miRNAs in exosomes (exo-miRs) can report the disease progression of organ injury. Using rat renal ischemia-reperfusion injury (IRI) as a model of acute kidney injury (AKI), we determined temporally-released exo-miRs in urine during IRI and found that these exo-miRs could reliably mirror the progression of AKI. From the longitudinal measurements of miRNA expression in kidney and urine, we found that release of exo- miRs was a regulated sorting process. In the injury state, miR-16, miR-24, and miR-200c were increased in the urine. Interestingly, expression of target mRNAs of these exo-miRs was significantly altered in renal medulla. Next, in the early recovery state, exo-miRs (miR-9a, miR-141, miR-200a, miR-200c, miR-429), which share Zeb1/2 as a common target mRNA, were upregulated together, indicating that they reflect TGF-ß-associated renal fibrosis. Finally, release of exo-miRs (miR-125a, miR-351) was regulated by TGF-ß1 and was able to differentiate the sham and IRI even after the injured kidneys were recovered. Altogether, these data indicate that exo-miRs released in renal IRI are associated with TGF-ß signaling. Temporal release of exo-miRs which share targets might be a regulatory mechanism to control the progression of AKI.


Assuntos
Injúria Renal Aguda/diagnóstico , Exossomos/genética , Medula Renal/fisiologia , MicroRNAs/genética , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Progressão da Doença , Fibrose , Perfilação da Expressão Gênica , Humanos , Medula Renal/patologia , Masculino , MicroRNAs/análise , Técnicas de Diagnóstico Molecular , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Urina/química
8.
Acta Physiol (Oxf) ; 226(3): e13266, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30770642

RESUMO

AIMS: In patients with essential hypertension, abnormal renal sodium handling includes exaggerated natriuresis in response to extracellular volume expansion. We tested the hypothesis that exaggerated natriuresis is associated with increases in medullary and/or cortical renal blood flow. METHODS: Patients with mild essential hypertension, but no signs of end organ damage, and control subjects were studied after 4 days of dietary standardization (<60 mmol Na+  day-1 ) preceded in patients by a 14-day drug washout period. On the study day, subjects received a 4-hour intravenous volume expansion with saline (2.1% of body mass). Renal medullary and cortical blood flows were measured by PET scanning using H215 O as tracer; anatomical regions of interest were defined by contrast-enhanced CT scanning. RESULTS: In patients, arterial blood pressure increased during volume expansion (107 ± 2-114 ± 3 mm Hg, P < 0.05) in contrast to the control group (92 ± 2-92 ± 2 mm Hg). Renal sodium excretion increased more in patients than in controls (+133 ± 31 µmol min-1 vs +61 ± 14 µmol min-1 , respectively, P < 0.05) confirming exaggerated natriuresis. During volume expansion, renal medullary blood flow did not change significantly in patients (2.8 ± 0.4-2.5 ± 0.5 mL (g tissue)-1  min-1 ) or in controls (3.2 ± 0.3-3.1 ± 0.2 mL (g tissue)-1 min-1 ). In control subjects, renal cortical blood flow fell during volume expansion (4.1 ± 0.3-3.7 ± 0.2 mL (g tissue)-1  min-1 , P < 0.05) in contrast to patients in which deviations remained insignificant. CONCLUSION: Exaggerated natriuresis, a hallmark of essential hypertension, is not mediated by increases in regional, renal blood flow.


Assuntos
Hipertensão Essencial/fisiopatologia , Hipertensão/fisiopatologia , Natriurese/fisiologia , Circulação Renal/fisiologia , Adulto , Pressão Sanguínea/fisiologia , Feminino , Taxa de Filtração Glomerular/fisiologia , Hemodinâmica/fisiologia , Humanos , Medula Renal/fisiologia , Masculino , Pessoa de Meia-Idade , Fluxo Sanguíneo Regional/fisiologia
9.
Sci Rep ; 8(1): 14157, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242165

RESUMO

Nephrocalcinosis often begins on a calcium phosphate deposit, at the tip of the medullo-papillary complex (MPC) known as Randall's plaque (RP). Contextualizing proximally observed biominerals within the MPC has led us to postulate a mechanobiological switch that can trigger interstitial biomineralization at the MPC tip, remote from the intratubular biominerals. Micro X-ray computed tomography scans of human MPCs correlated with transmission and scanning electron micrographs, and X-ray energy dispersive spectrometry demonstrated novel findings about anatomically-specific biominerals. An abundance of proximal intratubular biominerals were associated with emergence of distal interstitial RP. The fundamental architecture of the MPC and mineral densities at the proximal and distal locations of the MPC differed markedly. A predominance of plate-like minerals or radially oriented plate-like crystallites within spheroidal minerals in the proximal intratubular locations, and core-shell type crystallites within spheroidal minerals in distal interstitial locations were observed. Based on the MPC anatomic location of structure-specific biominerals, a biological switch within the mineral-free zone occurring between the proximal and distal locations is postulated. The "on" and "off" switch is dependent on changes in the pressure differential resulting from changes in tubule diameters; the "Venturi effect" changes the "circumferential strain" and culminates in interstitial crystal deposits in the distal tubule wall in response to proximal tubular obstruction. These distal interstitial mineralizations can emerge into the collecting system of the kidney linking nephrocalcinosis with nephrolithiasis.


Assuntos
Biomineralização/fisiologia , Medula Renal/fisiologia , Fosfatos de Cálcio/metabolismo , Humanos , Medula Renal/metabolismo , Minerais/metabolismo , Nefrocalcinose/metabolismo , Nefrocalcinose/fisiopatologia , Microtomografia por Raio-X/métodos
10.
Physiol Rep ; 6(18): e13846, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230259

RESUMO

A long-term goal in renal physiology is to understand the mechanisms involved in collecting duct function and regulation at a cellular and molecular level. The first step in modeling of these mechanisms, which can provide a guide to experimentation, is the generation of a list of model components. We have curated a list of proteins expressed in the rat renal inner medullary collecting duct (IMCD) from proteomic data from 18 different publications. The database has been posted as a public resource at https://hpcwebapps.cit.nih.gov/ESBL/Database/IMCD_Proteome_Database/. It includes 8956 different proteins. To search the IMCD Proteomic Database efficiently, we have created a Java-based program called curated database Basic Local Alignment Search Tool (cdbBLAST), which uses the NCBI BLAST kernel to search for specific amino acid sequences corresponding to proteins in the database. cdbBLAST reports information on the matched protein and identifies proteins in the database that have similar sequences. We have also adapted cdbBLAST to interrogate our previously published IMCD Transcriptome Database. We have made the cdbBLAST program available for use either as a web application or a downloadable .jar file at https://hpcwebapps.cit.nih.gov/ESBL/Database/cdbBLAST/. Database searching based on protein sequence removes ambiguities arising from the standard search method based on official gene symbols and allows the user efficient identification of related proteins that may fulfill the same functional roles.


Assuntos
Bases de Dados Genéticas , Medula Renal/fisiologia , Proteoma/genética , Transcriptoma/genética , Sequência de Aminoácidos , Animais , Ratos
11.
J Comp Physiol B ; 188(6): 899-918, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29797052

RESUMO

Mammalian kidneys play an essential role in balancing internal water and salt concentrations. When water needs to be conserved, the renal medulla produces concentrated urine. Central to this process of urine concentration is an osmotic gradient that increases from the corticomedullary boundary to the inner medullary tip. How this gradient is generated and maintained has been the subject of study since the 1940s. While it is generally accepted that the outer medulla contributes to the gradient by means of an active process involving countercurrent multiplication, the source of the gradient in the inner medulla is unclear. The last two decades have witnessed advances in our understanding of the urine-concentrating mechanism. Details of medullary architecture and permeability properties of the tubules and vessels suggest that the functional and anatomic relationships of these structures may contribute to the osmotic gradient necessary to concentrate urine. Additionally, we are learning more about the membrane transporters involved and their regulatory mechanisms. The role of medullary architecture and membrane transporters in the mammalian urine-concentrating mechanism are the focus of this review.


Assuntos
Medula Renal/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Urina , Animais , Humanos , Medula Renal/anatomia & histologia
12.
Physiol Genomics ; 50(6): 440-447, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29602296

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Inflamação/genética , Medula Renal/fisiologia , Medula Renal/fisiopatologia , Redes e Vias Metabólicas/genética , Perfusão , Animais , Teorema de Bayes , Respiração Celular , Hipertensão/genética , Metaboloma , Metabolômica , Mitocôndrias/metabolismo , Ratos Endogâmicos Dahl , Análise de Regressão , Software
13.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29237738

RESUMO

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.


Assuntos
Angiopoietina-1/fisiologia , Angiopoietina-2/fisiologia , Líquido Extracelular/metabolismo , Capacidade de Concentração Renal/fisiologia , Medula Renal/irrigação sanguínea , Receptor TIE-2/fisiologia , Angiopoietina-1/deficiência , Angiopoietina-1/genética , Angiopoietina-2/deficiência , Angiopoietina-2/genética , Animais , Padronização Corporal , Linhagem da Célula , Endotélio Vascular , Genes Reporter , Idade Gestacional , Proteínas de Homeodomínio/análise , Doenças Renais Císticas/genética , Medula Renal/embriologia , Medula Renal/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miofibroblastos/patologia , Osmose , Receptor TIE-2/deficiência , Receptor TIE-2/genética , Circulação Renal , Transdução de Sinais , Proteínas Supressoras de Tumor/análise , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/análise
14.
Physiol Rep ; 5(12)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28646097

RESUMO

To learn more about controlling renal interstitial hydrostatic pressure (RIHP), we assessed its response to renal medullary direct interstitial volume expansion (rmDIVE = 100 µL bolus infusion/30 sec). Three experimental series (S) were performed in hydropenic, anesthetized, right-nephrectomized, acute left renal-denervated and renal perfusion pressure-controlled rats randomly assigned to groups in each S. S1: Rats without hormonal clamp were contrasted before and after rmDIVE induced via 0.9% saline solution bolus (SS group) or 2% albumin in SS bolus (2% ALB + SS group). Subcapsular ΔRIHP rose slowly, progressively and similarly in both groups by ~3 mmHg. S2: Rats under hormonal clamp were contrasted before and after sham rmDIVE (time CTR group) and real rmDIVE induced via either SS bolus (SS group) or SS bolus containing the subcutaneous tissue fibroblast relaxant dibutyryl-cAMP (SS + db-cAMP group). ΔRIHP showed time, group, and time*group interaction effects with a biphasic response (early: ~1 mmHg; late: ~4 mmHg) in the SS group that was absent in the SS + db-cAMP group. S3: Two groups of rats (SS and SS + db-cAMP) under hormonal clamp were contrasted as in S2, producing similar ΔRIHP results to those of S2 but showing a slow, progressive, and indistinct decrease in renal outer medullary blood flow in both groups. These results provide highly suggestive preliminary evidence that the renal interstitium is capable of contracting reactively in vivo in response to rmDIVE with SS and demonstrate that such a response is abolished when db-cAMP is interstitially and concomitantly infused.


Assuntos
Pressão Hidrostática , Medula Renal/fisiologia , Animais , Bucladesina/farmacologia , Fibroblastos/efeitos dos fármacos , Medula Renal/citologia , Medula Renal/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Cloreto de Sódio/farmacologia
15.
Nephron ; 133(3): 183-92, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27362585

RESUMO

AIMS: Arterial spin labelling (ASL) MRI measures perfusion without administration of contrast agent. While ASL has been validated in animals and healthy volunteers (HVs), application to chronic kidney disease (CKD) has been limited. We investigated the utility of ASL MRI in patients with CKD. METHODS: We studied renal perfusion in 24 HVs and 17 patients with CKD (age 22-77 years, 40% male) using ASL MRI at 3.0T. Kidney function was determined using estimated glomerular filtration rate (eGFR). T1 relaxation time was measured using modified look-locker inversion and xFB02;ow-sensitive alternating inversion recovery true-fast imaging and steady precession was performed to measure cortical and whole kidney perfusion. RESULTS: T1 was higher in CKD within cortex and whole kidney, and there was association between T1 time and eGFR. No association was seen between kidney size and volume and either T1, or ASL perfusion. Perfusion was lower in CKD in cortex (136 ± 37 vs. 279 ± 69 ml/min/100 g; p < 0.001) and whole kidney (146 ± 24 vs. 221 ± 38 ml/min/100 g; p < 0.001). There was significant, negative, association between T1 longitudinal relaxation time and ASL perfusion in both the cortex (r = -0.75, p < 0.001) and whole kidney (r = -0.50, p < 0.001). There was correlation between eGFR and both cortical (r = 0.73, p < 0.01) and whole kidney (r = 0.69, p < 0.01) perfusion. CONCLUSIONS: Significant differences in renal structure and function were demonstrated using ASL MRI. T1 may be representative of structural changes associated with CKD; however, further investigation is required into the pathological correlates of reduced ASL perfusion and increased T1 time in CKD.


Assuntos
Córtex Renal/diagnóstico por imagem , Falência Renal Crônica/diagnóstico por imagem , Medula Renal/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Córtex Renal/fisiologia , Córtex Renal/fisiopatologia , Medula Renal/fisiologia , Medula Renal/fisiopatologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
16.
Sci Rep ; 6: 26251, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27185292

RESUMO

Classic methods for delivery of agents to specific organs are technically challenging and causes superfluous stress. The current study describes a method using programmable, implantable peristaltic pumps to chronically deliver drugs in vivo, while allowing animals to remain undisturbed for accurate physiological measurements. In this study, two protocols were used to demonstrate accurate drug delivery to the renal medulla. First, the vasopressin receptor-2 agonist, dDAVP, was delivered to the renal medulla resulting in a significant increase in water retention, urine osmolality and aquaporin-2 expression and phosphorylation. Second, in a separate group of rats, the histone deacetylase (HDAC) inhibitor, MS275, was delivered to the renal medulla. HDAC inhibition resulted in a significant increase in histone H3-acetylation, the hallmark for histone deacetylase inhibition. However, this was confined to the medulla, as the histone H3-acetylation was similar in the cortex of vehicle and MS275 infused rats, suggesting targeted drug delivery without systemic spillover. Thus, implantable, peristaltic pumps provide a number of benefits compared to externalized chronic catheters and confer specific delivery to target organs.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Bombas de Infusão Implantáveis , Acetilação/efeitos dos fármacos , Animais , Antidiuréticos/administração & dosagem , Aquaporina 2/metabolismo , Benzamidas/administração & dosagem , Desamino Arginina Vasopressina/administração & dosagem , Desenho de Equipamento , Inibidores de Histona Desacetilases/administração & dosagem , Histonas/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/fisiologia , Masculino , Especificidade de Órgãos , Concentração Osmolar , Piridinas/administração & dosagem , Ratos , Ratos Sprague-Dawley
17.
Life Sci ; 159: 144-147, 2016 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26724217

RESUMO

AIMS: Endothelin (ET)-1 promotes natriuresis via the endothelin B receptor (ETB) within the renal medulla. In male rats, direct interstitial infusion of ET-1 into the renal medulla has no effect on renal sodium and water excretion but is associated with endothelin A receptor (ETA)-dependent reductions in medullary blood flow. Loss of ETB function leads to salt-sensitive hypertension. We hypothesized that HS intake would increase the natriuretic and diuretic response to renal medullary infusion of ET peptides. MAIN METHODS: Male Sprague-Dawley (SD) rats were fed a normal (NS) or high (HS) salt diet for 7days. Rats were anesthetized and a catheter implanted in the renal medulla for interstitial infusion along with a ureteral catheter for urine collection. Medullary infusion of a low dose of ETB receptor agonist, sarafotoxin 6c (S6c; 0.15µg/kg/h), or ET-1 (0.45µg/kg/h) was used to determine changes in sodium excretion (UNaV). KEY FINDINGS: In HS fed rats, intramedullary infusion of a low dose of S6c induced a significant increase in UNaV, roughly 2-fold over baseline, compared to no response to this low dose in NS fed rats. In HS fed rats, intramedullary infusion of ET-1 induced a significantly greater increase in UNaV compared to NS fed rats, although this increase was not different from the HS time control studies. SIGNIFICANCE: We conclude that high salt intake enhances the diuretic and natriuretic effects of ETB receptor activation in vivo consistent with a role for the ETB receptor in maintaining fluid-electrolyte homeostasis.


Assuntos
Medula Renal/efeitos dos fármacos , Receptor de Endotelina B/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Animais , Medula Renal/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina B/fisiologia
18.
PLoS One ; 10(8): e0134477, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26280830

RESUMO

The kidney is one of the main organs that produces ammonia and release it into the circulation. Under normal conditions, between 30 and 50% of the ammonia produced in the kidney is excreted in the urine, the rest being absorbed into the systemic circulation via the renal vein. In acidosis and in some pathological conditions, the proportion of urinary excretion can increase to 70% of the ammonia produced in the kidney. Mechanisms regulating the balance between urinary excretion and renal vein release are not fully understood. We developed a mathematical model that reflects current thinking about renal ammonia handling in order to investigate the role of each tubular segment and identify some of the components which might control this balance. The model treats the movements of water, sodium chloride, urea, NH3 and [Formula: see text], and non-reabsorbable solute in an idealized renal medulla of the rat at steady state. A parameter study was performed to identify the transport parameters and microenvironmental conditions that most affect the rate of urinary ammonia excretion. Our results suggest that urinary ammonia excretion is mainly determined by those parameters that affect ammonia recycling in the loops of Henle. In particular, our results suggest a critical role for interstitial pH in the outer medulla and for luminal pH along the inner medullary collecting ducts.


Assuntos
Amônia/urina , Medula Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Alça do Néfron/fisiologia , Modelos Biológicos , Algoritmos , Compostos de Amônio/análise , Animais , Simulação por Computador , Concentração de Íons de Hidrogênio , Ratos , Cloreto de Sódio/análise , Ureia/análise , Água/análise
19.
Am J Physiol Renal Physiol ; 309(7): F627-37, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26290371

RESUMO

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.


Assuntos
Medula Renal/anatomia & histologia , Medula Renal/fisiologia , Aquaporina 1/metabolismo , Capilares/metabolismo , Epitélio/metabolismo , Humanos , Imageamento Tridimensional , Imuno-Histoquímica , Técnicas In Vitro , Capacidade de Concentração Renal/fisiologia , Túbulos Renais/metabolismo , Túbulos Renais Coletores/metabolismo , Consumo de Oxigênio
20.
Kidney Int ; 88(5): 1079-87, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26108066

RESUMO

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.


Assuntos
Córtex Renal/irrigação sanguínea , Medula Renal/irrigação sanguínea , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Circulação Renal/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Endotélio Vascular/metabolismo , Expressão Gênica/efeitos dos fármacos , Córtex Renal/fisiologia , Medula Renal/fisiologia , Masculino , Natriurese/efeitos dos fármacos , Óxido Nítrico/metabolismo , Oxigênio/sangue , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores Purinérgicos P2X7/genética , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...