Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
J Physiol ; 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39057844

ABSTRACT

Renal ischaemia and reperfusion (I/R) is caused by a sudden temporary impairment of the blood flow. I/R is a prevalent cause of acute kidney injury. As nitric oxide generated by inducible nitric oxide synthase (iNOS) has detrimental effects during I/R, the pharmacological blockade of iNOS has been proposed as a potential strategy to prevent I/R injury. The aim of this study was to improve the understanding of 1400W (an iNOS inhibitor) on renal I/R as a pharmacological strategy against kidney disease. BALB/c mice received 30 min of bilateral ischaemia, followed by 48 h or 28 days of reperfusion. Vehicle or 1400W (10 mg/kg) was administered 30 min before inducing ischaemia. We found that after 48 h of reperfusion 1400W decreased the serum creatinine, blood urea nitrogen, neutrophil gelatinase-associated lipocalin and proliferating cell nuclear antigen 3 in the I/R animals. Unexpectedly, we observed mRNA upregulation of genes involved in kidney injury, cell-cycle arrest, inflammation, mesenchymal transition and endothelial activation in the renal medulla of sham animals treated with 1400W. We also explored if 1400W promoted chronic kidney dysfunction 28 days after I/R and did not find significant alterations in renal function, fibrosis, blood pressure or mortality. The results provide evidence that 1400W may have adverse effects in the renal medulla. Importantly, our data point to 1400W-induced endothelial dysfunction, establishing therapeutic limitations for its use. KEY POINTS: Acute kidney injury is a global health problem associated with high morbidity and mortality. The pharmacological blockade of inducible nitric oxide synthase (iNOS) has been proposed as a potential strategy to prevent AKI induced by ischaemia and reperfusion (I/R). Our main finding is that 1400W, a selective and irreversible iNOS inhibitor with low toxicity that is proposed as a therapeutic strategy to prevent kidney I/R injury, produces aberrant gene expression in the medulla associated to tissue injury, cell cycle arrest, inflammation, mesenchymal transition and endothelial activation. The negative effect of 1400W observed in the renal medulla at 48 h from drug administration, is transient as it did not translate into a chronic kidney disease condition.

2.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39337519

ABSTRACT

The Mongolian sheep, emblematic of the Inner Mongolian grasslands, is renowned for its exceptional stress resistance and adaptability to harsh environments, drawing considerable attention. Recent research has unveiled the novel role of γ-aminobutyric acid (GABA) in combating oxidative stress. This investigation examined how GABA impacts renal-cortex and medulla cells from Mongolian sheep exposed to high-glucose stress conditions, utilizing gene expression analysis and non-targeted metabolomics. Elevated glucose levels significantly reduced the viability of Mongolian sheep renal cells and increased reactive oxygen species (ROS) levels. Conversely, the introduction of GABA notably enhanced cell viability, reduced ROS production, and stimulated the expression of antioxidant genes (e.g., Gpx, SOD, CAT) in the renal cortex. In the renal medulla, CAT expression increased, while Gpx gene expression showed mixed responses. Metabolomics analysis indicated that high-glucose exposure altered various metabolites, whereas GABA alleviated the metabolic stress induced by high glucose through modulating glycolysis and the tricarboxylic acid cycle. In Mongolian sheep renal cells, GABA effectively mitigated oxidative damage triggered by high-glucose stress by upregulating antioxidant genes and regulating metabolic pathways, revealing insights into its potential mechanism for adapting to extreme environments. This finding offers a fresh perspective on understanding the stress resilience of Mongolian sheep and may provide valuable insights for research across diverse disciplines.


Subject(s)
Glucose , Oxidative Stress , Reactive Oxygen Species , gamma-Aminobutyric Acid , Animals , Oxidative Stress/drug effects , Glucose/metabolism , gamma-Aminobutyric Acid/metabolism , Sheep , Reactive Oxygen Species/metabolism , Kidney/metabolism , Kidney/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Cell Survival/drug effects , Cells, Cultured , Metabolomics/methods
3.
Nitric Oxide ; 125-126: 40-46, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35700961

ABSTRACT

Hypertension significantly contributes to overall morbidity and mortality worldwide, and animal models of hypertension provide important tools to verify the physiological and molecular mechanisms underlying the development of the disease. A review of the most important models available would provide an insight into the appropriate targets to be addressed in the treatment of different forms of human hypertension. In the animal models discussed a special attention is given to the status and pathophysiological role of nitric oxide and its interaction with reactive oxygen species and oxidative stress. Another focus of the review are the processes running in the renal medulla which are still insufficiently explored. Deficient nitric oxide synthesis and its reduced bioavailability are important determinants of hypertension since NO is recognized as a major control factor of vascular tone homeostasis. For decades perfusion of the renal medulla has also been regarded as one of the blood pressure control factors and, noteworthily, the renal medulla belongs to the tissues with the highest NO content. The list of most often applied animal hypertension models reviewed here includes variants of salt-induced hypertension, the models with genetic background: such as spontaneously hypertensive rats (SHR) and Dahl salt sensitive (SS/SR) rats, Goldblatt 2K-1C hypertensive rats, and also the pharmacologically-plus-dietary salt-induced model known as DOCA-salt hypertension.


Subject(s)
Hypertension , Nitric Oxide , Animals , Blood Pressure , Humans , Models, Animal , Oxidative Stress/physiology , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary
4.
Br J Anaesth ; 128(6): 931-948, 2022 06.
Article in English | MEDLINE | ID: mdl-35465952

ABSTRACT

Perioperative hypotension is common and associated with poor outcomes, including acute kidney injury (AKI). The mechanistic link between perioperative hypotension and AKI is at least partly a consequence of the susceptibility of the kidney, and particularly the renal medulla, to ischaemia and hypoxia. Several critical gaps in our knowledge lead to uncertainty about when and how to intervene to prevent AKI attributable to perioperative hypotension. First, although we know that the risk of AKI varies with both the severity and duration of hypotensive episodes, 'safe' levels of arterial pressure have not been identified. Second, there have been few adequately powered clinical trials of interventions to avoid perioperative hypotension. Thus, most evidence surrounding perioperative hypotension is observational rather than based on randomised clinical trials. This means that the link between perioperative hypotension and AKI may represent association (where both phenomena reflect illness severity) rather than causation. Third, there is little information regarding the relative risks and benefits of various clinically available therapies (e.g. vasoconstrictors, i.v. fluids, or both) to treat and prevent perioperative hypotension, particularly with regard to renal medullary perfusion and oxygenation. Fourth, there are currently no validated, clinically feasible methods for real-time clinical monitoring of renal perfusion or oxygenation. Thus, future developments in perioperative kidney-protective strategies must rely on the development of methods to better monitor renal perfusion and oxygenation in the perioperative period, and thereby guide timing, intensity, type, and duration of interventions.


Subject(s)
Acute Kidney Injury , Hypotension , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Arterial Pressure , Humans , Hypotension/etiology , Hypotension/prevention & control , Kidney , Postoperative Complications/drug therapy , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Vasoconstrictor Agents/therapeutic use
5.
Clin Exp Pharmacol Physiol ; 49(2): 228-241, 2022 02.
Article in English | MEDLINE | ID: mdl-34674291

ABSTRACT

Acute kidney injury (AKI) is a common and serious post-operative complication of cardiac surgery. The value of a predictive biomarker is determined not only by its predictive efficacy, but also by how early this prediction can be made. For a biomarker of cardiac surgery-associated AKI, this is ideally during the intra-operative period. Therefore, in 82 adult patients undergoing cardiac surgery requiring cardiopulmonary bypass (CPB), we prospectively compared the predictive efficacy of various blood and urinary biomarkers with that of continuous measurement of urinary oxygen tension (UPO2 ) at pre-determined intra- and post-operative time-points. None of the blood or urine biomarkers we studied showed predictive efficacy for post-operative AKI when measured intra-operatively. When treated as a binary variable (≤ or > median for the whole cohort), the earliest excess risk of AKI was predicted by an increase in urinary neutrophil gelatinase-associated lipocalin (NGAL) at 3 h after entry into the intensive care unit (odds ratio [95% confidence limits], 2.86 [1.14-7.21], p = 0.03). Corresponding time-points were 6 h for serum creatinine (3.59 [1.40-9.20], p = 0.008), and 24 h for plasma NGAL (4.54 [1.73-11.90], p = 0.002) and serum cystatin C (6.38 [2.35-17.27], p = 0.001). In contrast, indices of intra-operative urinary hypoxia predicted AKI after weaning from CPB, and in the case of a fall in UPO2 to ≤10 mmHg, during the rewarming phase of CPB (3.00 [1.19-7.56], p = 0.02). We conclude that continuous measurement of UPO2 predicts AKI earlier than plasma or urinary NGAL, serum cystatin C, or early post-operative changes in serum creatinine.


Subject(s)
Acute Kidney Injury , Cardiac Surgical Procedures , Acute Kidney Injury/diagnosis , Acute Kidney Injury/etiology , Acute-Phase Proteins , Adult , Biomarkers , Cardiac Surgical Procedures/adverse effects , Creatinine , Humans , Lipocalins , Oxygen , Predictive Value of Tests , Proto-Oncogene Proteins
6.
Int J Mol Sci ; 23(11)2022 Jun 03.
Article in English | MEDLINE | ID: mdl-35682969

ABSTRACT

Pregnancy is characterized by adaptations in the function of several maternal body systems that ensure the development of the fetus whilst maintaining health of the mother. The renal system is responsible for water and electrolyte balance, as well as waste removal. Thus, it is imperative that structural and functional changes occur in the kidney during pregnancy. However, our knowledge of the precise morphological and molecular mechanisms occurring in the kidney during pregnancy is still very limited. Here, we investigated the changes occurring in the mouse kidney during pregnancy by performing an integrated analysis involving histology, gene and protein expression assays, mass spectrometry profiling and bioinformatics. Data from non-pregnant and pregnant mice were used to identify critical signalling pathways mediating changes in the maternal kidneys. We observed an expansion of renal medulla due to proliferation and infiltration of interstitial cellular constituents, as well as alterations in the activity of key cellular signalling pathways (e.g., AKT, AMPK and MAPKs) and genes involved in cell growth/metabolism (e.g., Cdc6, Foxm1 and Rb1) in the kidneys during pregnancy. We also generated plasma and urine proteomic profiles, identifying unique proteins in pregnancy. These proteins could be used to monitor and study potential mechanisms of renal adaptations during pregnancy and disease.


Subject(s)
Kidney , Proteomics , Animals , Female , Fetus/metabolism , Kidney/metabolism , Kidney Medulla/metabolism , Mice , Pregnancy , Proteins/metabolism , Water-Electrolyte Balance
7.
Am J Physiol Renal Physiol ; 320(6): F1059-F1079, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33779315

ABSTRACT

Ammonia generated within the kidney is partitioned into a urinary fraction (the key buffer for net acid excretion) and an aliquot delivered to the systemic circulation. The physiology of this partitioning has yet to be examined in a kidney model, and that was undertaken in this work. This involves explicit representation of the cortical labyrinth, so that cortical interstitial solute concentrations are computed rather than assigned. A detailed representation of cortical vasculature has been avoided by making the assumption that solute concentrations within the interstitium and peritubular capillaries are likely to be identical and that there is little to no modification of venous composition as blood flows to the renal vein. The model medullary ray has also been revised to include a segment of proximal straight tubule, which supplies ammonia to this region. The principal finding of this work is that cortical labyrinth interstitial ammonia concentration is likely to be several fold higher than systemic arterial ammonia. This elevation of interstitial ammonia enhances ammonia secretion in both the proximal convoluted tubule and distal convoluted tubule, with uptake by Na+-K+-ATPases of both segments. Model prediction of urinary ammonia excretion was concordant with measured values, but at the expense of greater ammoniagenesis, with high rates of renal venous ammonia flux. This derives from a limited capability of the model medulla to replicate the high interstitial ammonia concentrations that are required to drive collecting duct ammonia secretion. Thus, renal medullary ammonia trapping appears key to diverting ammonia from the renal vein to urine, but capturing the underlying physiology remains a challenge.NEW & NOTEWORTHY This is the first mathematical model to estimate solute concentrations within the kidney cortex. The model predicts cortical ammonia to be several fold greater than in the systemic circulation. This higher concentration drives ammonia secretion in proximal and distal tubules. The model reveals a gap in our understanding of how ammonia generated within the cortex is channeled efficiently into the final urine.


Subject(s)
Ammonia/metabolism , Kidney/physiology , Models, Biological , Ammonia/urine , Animals , Biological Transport , Kidney/blood supply , Rats
8.
Genomics ; 112(3): 2633-2639, 2020 05.
Article in English | MEDLINE | ID: mdl-32070763

ABSTRACT

Camels as a sort of animal long living in desert have evolved stress-resistance characteristics to adapt to environment with high temperature and water shortage environment. However, the research of non-coding RNA (ncRNA)-mediated molecular regulation about how camel responds to arid condition in post-transcriptional regulation level is deficient. Under water-deprivation stress, by RNA-sequencing of camel renal medulla associated with regulating water metabolism, we detected significantly differential 575 alternative splicing events (ASEs) and 17 mRNAs, 26 miRNAs and 0 lncRNA. The down-regulated ACLY and LOC105061856, up-regulated PCBP2 and miR-195 potentially targeting LOC105061856 and PCBP2 mRNA were selected as candidate resistance-related genes. In quantitative experiment, the expression level of above four genes was consistent with RNA-seq data by qRT-PCR. The suppressive cell dehydration with down-regulated ACLY, inhibitive aerobic respiration with down-regulated LOC105061856 targeted by miR-195 and strong anti-oxidative capability with PCBP2 aimed by miR-195 may be regulatory modes of camel renal medulla adapting to water-deprivation condition.


Subject(s)
Camelus/genetics , Gene Expression Regulation/genetics , Kidney Medulla/metabolism , Alternative Splicing , Animals , Camelus/metabolism , Dehydration/genetics , Dehydration/metabolism , Dehydration/veterinary , Droughts , Female , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism
9.
Clin Exp Pharmacol Physiol ; 47(8): 1323-1332, 2020 08.
Article in English | MEDLINE | ID: mdl-32163610

ABSTRACT

In the context of the ongoing debate on the mechanism of blood pressure (BP) regulation and pathophysiology of arterial hypertension ("renocentric" vs "neural" concepts), attention is focused on the putative regulatory role of changes in renal medullary blood flow (MBF). Experimental evidence is analysed with regard to the question whether an elevation of BP and renal perfusion pressure (RPP) is likely to increase MBF due to its impaired autoregulation. It is concluded that such increases have been clearly documented only in rats with extracellular fluid volume expansion. A possible translation of this finding to BP regulation in health and hypertension in humans may only be a matter of speculation. Within the "renocentric" theory, the key event leading to restoration of initial BP level is pressure natriuresis. Its relation to elevation of renal interstitial hydrostatic pressure and to the phenomenon of "wash-out" of renal medullary solutes by increasing MBF is discussed. We also assessed the validity of data supporting the putative mechanism of short-term restoration of elevated BP owing to the release of a vasodilator lipid (medullipin) by the medulla. The structure of the proposed medullary lipid is still undefined, and there is no sound evidence on its mediatory role in lowering elevated BP level. In conclusion, MBF change can hardly be regarded as a crucial event in the regulation of BP: it can be involved in the control of sodium excretion and BP only in some circumstances, although its contributory role cannot be excluded.


Subject(s)
Blood Pressure , Renal Circulation , Sodium/metabolism , Animals , Lipid Metabolism , Male , Rats
10.
Nephrology (Carlton) ; 25(2): 116-124, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31170320

ABSTRACT

AIM: In humans, nephrogenesis ceases before birth, but the renal medulla compartment continues to develop after birth. We aim to evaluate the relative growth of different renal compartments in preterm babies compared with age-matched term babies, and explore the impact of premature birth on postnatal renal maturation, remodelling and possible long-term implications. METHODS: This retrospective study compared the renal ultrasonographic images between preterm babies and term infants. Ultrasound images were obtained at 32 weeks (preterm), 37 weeks and at 6 months of age. Kidney volume, length, renal cortex and medulla thickness were measured and compared between preterm and term babies. RESULTS: Preterm babies were lighter in body weight and shorter for crown-heel length at age-matched 37 weeks. All kidney growth parameters were also smaller compared with term babies. However, by 6 months of age kidney volume and length measurements were no longer significantly different between the two groups though preterm babies were still significantly lighter and shorter. The catch-up of the overall kidney growth in preterm babies was mainly attributed to the hypertrophic growth of the renal cortex while the postnatal renal medulla growth was disrupted. This trend continued as the renal cortical thickness became significantly larger while the medulla became smaller in preterm babies at 6 months of age, compared with age-matched term baby. CONCLUSIONS: In preterm babies, the renal cortical region undergoes accelerated growth after birth while the renal medulla growth lags behind. Further investigations will be necessary to determine whether this has a negative impact on renal function later in life.


Subject(s)
Gestational Age , Infant, Premature/growth & development , Kidney , Child Development , Female , Humans , Infant, Newborn , Kidney/diagnostic imaging , Kidney/growth & development , Kidney/pathology , Kidney/physiopathology , Male , Organ Size , Prognosis , Retrospective Studies , Ultrasonography/methods
11.
Kidney Int ; 95(6): 1338-1346, 2019 06.
Article in English | MEDLINE | ID: mdl-31005272

ABSTRACT

Renal medullary hypoxia may contribute to cardiac surgery-associated acute kidney injury (AKI). However, the effects of cardiopulmonary bypass (CPB) on medullary oxygenation are poorly understood. Here we tested whether CPB causes medullary hypoxia and whether medullary oxygenation during CPB can be improved by increasing pump flow or mean arterial pressure (MAP). Twelve sheep were instrumented to measure whole kidney, medullary, and cortical blood flow and oxygenation. Five days later, under isoflurane anesthesia, CPB was initiated at a pump flow of 80 mL kg-1min-1 and target MAP of 70 mm Hg. Pump flow was then set at 60 and 100 mL kg-1min-1, while MAP was maintained at approximately 70 mm Hg. MAP was then increased by vasopressor (metaraminol, 0.2-0.6 mg/min) infusion at a pump flow of 80 mL kg-1min-1. CPB at 80 mL kg-1min-1 reduced renal blood flow (RBF), -61% less than the conscious state, perfusion in the cortex (-44%) and medulla (-40%), and medullary Po2 from 43 to 27 mm Hg. Decreasing pump flow from 80 to 60 mL kg-1min-1 further decreased RBF (-16%) and medullary Po2 from 25 to 14 mm Hg. Increasing pump flow from 80 to 100 mL kg-1min-1 increased RBF (17%) and medullary Po2 from 20 to 29 mm Hg. Metaraminol (0.2 mg/min) increased MAP from 63 to 90 mm Hg, RBF (47%), and medullary Po2 from 19 to 39 mm Hg. Thus, the renal medulla is susceptible to hypoxia during CPB, but medullary oxygenation can be improved by increasing pump flow or increasing target MAP by infusion of metaraminol.


Subject(s)
Acute Kidney Injury/prevention & control , Cardiopulmonary Bypass/adverse effects , Kidney Medulla/blood supply , Postoperative Complications/prevention & control , Vasoconstrictor Agents/administration & dosage , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Arterial Pressure/drug effects , Cardiopulmonary Bypass/instrumentation , Cardiopulmonary Bypass/methods , Cell Hypoxia/drug effects , Disease Models, Animal , Female , Humans , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Kidney Medulla/pathology , Metaraminol/administration & dosage , Oxygen/metabolism , Postoperative Complications/etiology , Postoperative Complications/pathology , Renal Circulation/drug effects , Renal Circulation/physiology , Sheep
12.
MAGMA ; 31(3): 375-382, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29289980

ABSTRACT

OBJECTIVE: Low-energy shockwave (SW) therapy attenuates damage in the stenotic kidney (STK) caused by atherosclerotic renal artery stenosis (ARAS). We hypothesized that magnetic resonance elastography (MRE) would detect attenuation of fibrosis following SW in unilateral ARAS kidneys. MATERIALS AND METHODS: Domestic pigs were randomized to control, unilateral ARAS, and ARAS treated with 6 sessions of SW over 3 consecutive weeks (n = 7 each) starting after 3 weeks of ARAS or sham. Four weeks after SW treatment, renal fibrosis was evaluated with MRE in vivo or trichrome staining ex vivo. Blood pressure, single-kidney renal-blood-flow (RBF) and glomerular-filtration-rate (GFR) were assessed. RESULTS: MRE detected increased stiffness in the STK medulla (15.3 ± 2.1 vs. 10.1 ± 0.8 kPa, p < 0.05) that moderately correlated with severity of fibrosis (R2 = 0.501, p < 0.01), but did not identify mild STK cortical or contralateral kidney fibrosis. Trichrome staining showed that medullary fibrosis was increased in ARAS and alleviated by SW (10.4 ± 1.8% vs. 2.9 ± 0.2%, p < 0.01). SW slightly decreased blood pressure and normalized STK RBF and GFR in ARAS. In the contralateral kidney, SW reversed the increase in RBF and GFR. CONCLUSION: MRE might be a tool for noninvasive monitoring of medullary fibrosis in response to treatment in kidney disease.


Subject(s)
Atherosclerosis/diagnostic imaging , Elasticity Imaging Techniques , Ischemia/diagnostic imaging , Kidney/diagnostic imaging , Magnetic Resonance Imaging , Animals , Atherosclerosis/physiopathology , Constriction, Pathologic , Disease Models, Animal , Female , Fibrosis , Glomerular Filtration Rate , Ischemia/pathology , Kidney/pathology , Kidney Cortex/diagnostic imaging , Kidney Cortex/physiopathology , Renal Artery/diagnostic imaging , Renal Artery/physiopathology , Renal Artery Obstruction/diagnostic imaging , Renal Artery Obstruction/physiopathology , Renal Circulation , Sus scrofa , Swine
13.
Differentiation ; 94: 1-7, 2017.
Article in English | MEDLINE | ID: mdl-27923152

ABSTRACT

The Wnt5a null mouse is a complex developmental model which, among its several posterior-localized axis defects, exhibits multiple kidney phenotypes, including duplex kidney and loss of the medullary zone. We previously reported that ablation of Wnt5a in nascent mesoderm causes duplex kidney formation as a result of aberrant development of the nephric duct and abnormal extension of intermediate mesoderm. However, these mice also display a loss of the medullary region late in gestation. We have now genetically isolated duplex kidney formation from the medullary defect by specifically targeting the progenitors for both the ureteric bud and metanephric mesenchyme. The conditional mutants fail to form a normal renal medulla but no longer exhibit duplex kidney formation. Approximately 1/3 of the mutants develop hydronephrosis in the kidneys either uni- or bilaterally when using Dll1Cre. The abnormal kidney phenotype becomes prominent at E16.5, which approximates the time when urine production begins in the mouse embryonic kidney, and is associated with a dramatic increase in apoptosis only in mutant kidneys with hydronephrosis. Methylene blue dye injection and histologic examination reveal that aberrant cell death likely results from urine toxicity due to an abnormal ureter-bladder connection. This study shows that Wnt5a is not required for development of the renal medulla and that loss of the renal medullary region in the Wnt5a-deleted kidney is caused by an abnormal ureter-bladder connection.


Subject(s)
Cell Differentiation/genetics , Hydronephrosis/genetics , Kidney/growth & development , Wnt-5a Protein/genetics , Animals , Hydronephrosis/physiopathology , Kidney/physiopathology , Mice , Mice, Knockout , Morphogenesis/genetics , Signal Transduction/genetics , Ureter/abnormalities , Ureter/growth & development , Urinary Bladder/abnormalities , Urinary Bladder/growth & development
14.
Am J Physiol Renal Physiol ; 313(2): F361-F369, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28468962

ABSTRACT

We recently reported that natriuresis produced by renal medullary salt loading is dependent on endothelin (ET)-1 and purinergic (P2) receptors in male rats. Because sex differences in ET-1 and P2 signaling have been reported, we decided to test whether ovarian sex hormones regulate renal medullary ET-1 and P2-dependent natriuresis. The effect of medullary NaCl loading on Na+ excretion was determined in intact and ovariectomized (OVX) female Sprague-Dawley rats with and without ET-1 or P2 receptor antagonism. Isosmotic saline (284 mosmol/kgH2O) was infused in the renal medullary interstitium of anesthetized rats during a baseline urine collection period, followed by isosmotic or hyperosmotic saline (1,800 mosmol/kgH2O) infusion. Medullary NaCl loading significantly enhanced Na+ excretion in intact and OVX female rats. ETA+B or P2 receptor blockade did not attenuate the natriuretic effect of medullary NaCl loading in intact females, whereas ETA+B or P2 receptor blockade attenuated the natriuretic response to NaCl loading in OVX rats. Activation of medullary P2Y2 and P2Y4 receptors by UTP infusion had no significant effect in intact females but enhanced Na+ excretion in OVX rats. Combined ETA+B receptor blockade significantly inhibited the natriuretic response to UTP observed in OVX rats. These data demonstrate that medullary NaCl loading induces ET-1 and P2-independent natriuresis in intact females. In OVX, activation of medullary P2 receptors promotes ET-dependent natriuresis, suggesting that ovarian hormones may regulate the interplay between the renal ET-1 and P2 signaling systems to facilitate Na+ excretion.


Subject(s)
Endothelin-1/metabolism , Kidney Medulla/metabolism , Natriuresis , Ovariectomy , Receptors, Purinergic P2Y2/metabolism , Receptors, Purinergic P2/metabolism , Renal Elimination , Sodium/urine , Animals , Endothelin Receptor Antagonists/pharmacology , Endothelin-1/genetics , Female , Kidney Medulla/drug effects , Natriuresis/drug effects , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2Y2/drug effects , Renal Elimination/drug effects , Signal Transduction , Sodium Chloride/administration & dosage , Sodium Chloride/metabolism , Time Factors
15.
Am J Physiol Renal Physiol ; 313(5): F1068-F1076, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28768662

ABSTRACT

The renal medulla, considered critical for the regulation of salt and water balance and long-term blood pressure control, is enriched in anandamide and two of its major metabolizing enzymes, cyclooxygenase-2 (COX-2) and fatty acid amide hydrolase (FAAH). Infusion of anandamide (15, 30, and 60 nmol·min-1·kg-1) into the renal medulla of C57BL/6J mice stimulated diuresis and salt excretion in a COX-2- but not COX-1-dependent manner. To determine whether endogenous endocannabinoids in the renal medulla can elicit similar effects, the effects of intramedullary isopropyl dodecyl fluorophosphate (IDFP), which inhibits the two major endocannabinoid hydrolases, were studied. IDFP treatment increased the urine formation rate and sodium excretion in a COX-2- but not COX-1-dependent manner. Neither anandamide nor IDFP affected the glomerular filtration rate. Neither systemic (0.625 mg·kg-1·30 min-1 iv) nor intramedullary (15 nmol·min-1·kg-1·30 min-1) IDFP pretreatment before intramedullary anandamide (15-30 nmol·min-1·kg-1) strictly blocked effects of anandamide, suggesting that hydrolysis of anandamide was not necessary for its diuretic effect. Intramedullary IDFP had no effect on renal blood flow but stimulated renal medullary blood flow. The effects of IDFP on urine flow rate and medullary blood flow were FAAH-dependent as demonstrated using FAAH knockout mice. Analysis of mouse urinary PGE2 concentrations by HPLC-electrospray ionization tandem mass spectrometry showed that IDFP treatment decreased urinary PGE2 These data are consistent with a role of FAAH and endogenous anandamide acting through a COX-2-dependent metabolite to regulate diuresis and salt excretion in the mouse kidney.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Diuresis , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Arachidonic Acids/metabolism , Cyclooxygenase 2/metabolism , Diuresis/drug effects , Endocannabinoids/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Monoacylglycerol Lipases/metabolism , Natriuresis/drug effects , Natriuresis/physiology , Polyunsaturated Alkamides/metabolism , Renal Circulation/physiology
16.
Mol Biol (Mosk) ; 51(3): 442-446, 2017.
Article in Russian | MEDLINE | ID: mdl-28707660

ABSTRACT

The concentration of soluble epoxide hydrolase (sEH) protein was studied in renal medulla of adult rats from hypertensive ISIAH strain and normotensive WAG strain. The sEH is a key enzyme in metabolism of epoxyeicosatrienoic acids capable of activating endothelial NO-synthase and nitrogen oxide formation, and therefore being vasodilators. An increase in the sEH protein concentration (that we found) allows one to assume that the oxidative stress is increased in the renal medulla of hypertensive rats, and the bloodflow is decreased.


Subject(s)
Epoxide Hydrolases/biosynthesis , Oxidative Stress/genetics , Stress, Physiological/genetics , Animals , Blood Pressure , Disease Models, Animal , Epoxide Hydrolases/isolation & purification , Humans , Hypertension/enzymology , Hypertension/pathology , Kidney Medulla/enzymology , Kidney Medulla/pathology , Male , Nitric Oxide Synthase/genetics , Nitrogen Oxides/metabolism , Rats
17.
BMC Genet ; 17(Suppl 3): 151, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28105926

ABSTRACT

BACKGROUND: The changes in the renal function leading to a reduction of medullary blood flow can have a great impact on sodium and water homeostasis and on the long-term control of arterial blood pressure. The RNA-Seq approach was used for transcriptome profiling of the renal medulla from hypertensive ISIAH and normotensive WAG rats to uncover the genetic basis of the changes underlying the renal medulla function in the ISIAH rats being a model of the stress-sensitive arterial hypertension and to reveal the genes which possibly may contribute to the alterations in medullary blood flow. RESULTS: Multiple DEGs specifying the function of renal medulla in ISIAH rats were revealed. The group of DEGs described by Gene Ontology term 'oxidation reduction' was the most significantly enriched one. The other groups of DEGs related to response to external stimulus, response to hormone (endogenous) stimulus, response to stress, and homeostatic process provide the molecular basis for integrated responses to homeostasis disturbances in the renal medulla of the ISIAH rats. Several DEGs, which may modulate the renal medulla blood flow, were detected. The reduced transcription of Nos3 pointed to the possible reduction of the blood flow in the renal medulla of ISIAH rats. CONCLUSIONS: The generated data may be useful for comparison with those from different models of hypertension and for identifying the common molecular determinants contributing to disease manifestation, which may be potentially used as new pharmacological targets.


Subject(s)
Hypertension/genetics , Kidney Medulla/metabolism , Transcriptome , Animals , Blood Pressure , Databases, Genetic , Discriminant Analysis , Disease Models, Animal , Hypertension/etiology , Hypertension/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Principal Component Analysis , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Rats , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Stress, Physiological/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Am J Physiol Renal Physiol ; 308(2): F149-56, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25377910

ABSTRACT

Thick ascending limbs reabsorb 30% of the filtered NaCl load. Nitric oxide (NO) produced by NO synthase 3 (NOS3) inhibits NaCl transport by this segment. In contrast, chronic angiotensin II (ANG II) infusion increases net thick ascending limb transport. NOS3 activity is regulated by changes in expression and phosphorylation at threonine 495 (T495) and serine 1177 (S1177), inhibitory and stimulatory sites, respectively. We hypothesized that NO production by thick ascending limbs is impaired by chronic ANG II infusion, due to reduced NOS3 expression, increased phosphorylation of T495, and decreased phosphorylation of S1177. Rats were infused with 200 ng·kg(-1)·min(-1) ANG II or vehicle for 1 and 5 days. ANG II infusion for 5 days decreased NOS3 expression by 40 ± 12% (P < 0.007; n = 6) and increased T495 phosphorylation by 147 ± 26% (P < 0.008; n = 6). One-day ANG II infusion had no significant effect. NO production in response to endothelin-1 was blunted in thick ascending limbs from ANG II-infused animals [ANG II -0.01 ± 0.06 arbitrary fluorescence units (AFU)/min vs. 0.17 ± 0.02 AFU/min in controls; P < 0.01]. This was not due to reduced endothelin-1 receptor expression. Phosphatidylinositol 3,4,5-triphosphate (PIP3)-induced NO production was also reduced in ANG II-infused rats (ANG II -0.07 ± 0.06 vs. 0.13 ± 0.04 AFU/min in controls; P < 0.03), and this correlated with an impaired ability of PIP3 to increase S1177 phosphorylation. We conclude that in ANG II-induced hypertension NO production by thick ascending limbs is impaired due to decreased NOS3 expression and altered phosphorylation.


Subject(s)
Angiotensin II/metabolism , Loop of Henle/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Protein Kinase C/metabolism , Animals , Male , Phosphorylation , Rats, Sprague-Dawley
19.
Am J Physiol Renal Physiol ; 309(4): F300-4, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26062876

ABSTRACT

Outer medullary isolated descending vasa recta have proven to be experimentally tractable, and consequently much has been learned about outer medullary vasa recta endothelial transport, pericyte contractile mechanisms, and tubulovascular interactions. In contrast, inner medullary vasa recta have never been isolated from any species, and therefore isolated vasa recta function has never been subjected to in vitro quantitative evaluation. As we teased out inner medullary thin limbs of Henle's loops from the Munich-Wistar rat, we found that vasa recta could be isolated using similar protocols. We isolated ∼30 inner medullary vasa recta from 23 adult male Munich-Wistar rats and prepared them for brightfield or electron microscopy, gene expression analysis by RT-PCR, or isolated tubule microperfusion. Morphological characteristics include branching and nonbranching segments exhibiting a thin endothelium, axial surface filaments radiating outward giving vessels a hairy appearance, and attached interstitial cells. Electron microscopy shows multiple cells, tight junctions, and either continuous or fenestrated endothelia. Isolated vasa recta express genes encoding the urea transporter UT-B and/or the fenestral protein PV-1, genes expressed in descending or ascending vasa recta, respectively. The transepithelial NaCl permeability (383.3 ± 60.0 × 10(-5) cm/s, mean ± SE, n = 4) was determined in isolated perfused vasa recta. Future quantitative analyses of isolated inner medullary vasa recta should provide structural and functional details important for more fully understanding fluid and solute flows through the inner medulla and their associated regulatory pathways.


Subject(s)
Blood Vessels/physiology , Dissection/methods , In Vitro Techniques , Kidney Medulla/blood supply , Loop of Henle/blood supply , Perfusion/methods , Renal Circulation , Animals , Biomarkers/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Capillary Permeability , Gene Expression Regulation , Male , Microscopy, Electron , RNA, Messenger/metabolism , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
20.
Am J Physiol Cell Physiol ; 307(5): C442-54, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965592

ABSTRACT

High extracellular NaCl, such as in the renal medulla, can perturb and even kill cells, but cells mount protective responses that enable them to survive and function. Many high-NaCl-induced perturbations and protective responses are known, but the signaling pathways involved are less clear. Change in protein phosphorylation is a common mode of cell signaling, but there was no unbiased survey of protein phosphorylation in response to high NaCl. We used stable isotopic labeling of amino acids in cell culture coupled to mass spectrometry to identify changes in protein phosphorylation in human embryonic kidney (HEK 293) cells exposed to high NaCl. We reproducibly identify >8,000 unique phosphopeptides in 4 biological replicate samples with a 1% false discovery rate. High NaCl significantly changed phosphorylation of 253 proteins. Western analysis and targeted ion selection mass spectrometry confirm a representative sample of the phosphorylation events. We analyze the affected proteins by functional category to infer how altered protein phosphorylation might signal cellular responses to high NaCl, including alteration of cell cycle, cyto/nucleoskeletal organization, DNA double-strand breaks, transcription, proteostasis, metabolism of mRNA, and cell death.


Subject(s)
Extracellular Fluid/metabolism , Membrane Proteins/metabolism , Sodium Chloride/toxicity , Chromatography, Liquid , Extracellular Fluid/drug effects , HEK293 Cells , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Chloride/chemistry , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL