Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Hypertension ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38841853

ABSTRACT

BACKGROUND: Clinical trials of renal denervation for the treatment of hypertension have shown a variety of off-target improvements in conditions associated with sympathetic overactivity. This may be due to the ablation of sympathoexcitatory afferent renal nerves, which are overactive under conditions of renal inflammation. Renal IL (interleukin)-1ß is elevated in the deoxycorticosterone acetate-salt model of hypertension, and its activity may be responsible for the elevation in afferent renal nerve activity and arterial pressure. METHODS: Continuous blood pressure recording of deoxycorticosterone acetate-salt mice with IL-1R (IL-1 receptor) knockout or antagonism was used individually and combined with afferent renal denervation (ARDN) to assess mechanistic overlap. Protein quantification and histological analysis of kidneys were performed to characterize renal inflammation. RESULTS: ARDN attenuated deoxycorticosterone acetate-salt hypertension (-20±2-Δmm Hg mean arterial pressure [MAP] relative to control at study end) to a similar degree as total renal denervation (-21±2-Δmm Hg MAP), IL-1R knockout (-16±4-Δmm Hg MAP), or IL-1R antagonism (-20±3-Δmm Hg MAP). The combination of ARDN with knockout (-18±2-Δmm Hg MAP) or antagonism (-19±4-Δmm Hg MAP) did not attenuate hypertension any further than ARDN alone. IL-1R antagonism was found to have an acute depressor effect (-15±3-Δmm Hg MAP, day 10) in animals with intact renal nerves but not those with ARDN. CONCLUSIONS: These findings suggest that IL-1R signaling is partially responsible for the elevated afferent renal nerve activity, which stimulates central sympathetic outflow to drive deoxycorticosterone acetate-salt hypertension.

2.
Am J Physiol Renal Physiol ; 326(1): F95-F104, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37916287

ABSTRACT

In the current study, we took advantage of the loss of protection from hypertension in SSCD247-/- rats to characterize the pathological effects of renal T-cells in isolation from the confounding effects of elevated renal perfusion pressure. Male SSCD247-/- and SSCD247+/+ littermates were fed 4.0% NaCl (high salt) diet to induce hypertension. Blood pressure was assessed continuously throughout the time course with radiotelemetry. Urine albumin and protein excretion were assessed on the final day of high salt. Renal injury and medullary transcriptome were assessed after completion of the high salt protocol. In contrast to previous studies, mean arterial pressure was not significantly different between SSCD247-/- and SSCD247+/+ rats. Despite this lack of pressure difference, urinary albumin was significantly lower in SSCD247-/- rats than their wild-type littermates. In the outer medulla, substantially more transcriptomic changes were found to correlate with endpoint blood pressure than with the absence of presence of renal T-cells. We also demonstrated that renal histological damage was driven by elevated renal perfusion pressure rather than the presence of renal T-cells. In conclusion, using the loss of protection from hypertension in SSCD247-/- rats, we demonstrated that renal perfusion pressure has more profound pathological effects on the kidney than renal T-cells. However, renal T-cells, independently of blood pressure, modulate the progression of albuminuria.NEW & NOTEWORTHY In vivo studies in a T-cell-deficient rat model of salt-sensitive hypertension (SSCD247-/- rats) were used to evaluate the role of T-cells on the development of hypertension and renal damage. Detailed physiological and transcriptomic analysis demonstrated no difference in blood pressure between rats with (SSCD247+/+) or without (SSCD247-/-) T-cells. Despite this, albuminuria was significantly lower in SSCD247-/- rats than SSCD247+/+ rats.


Subject(s)
Hypertension , Transcriptome , Rats , Male , Animals , Albuminuria/metabolism , T-Lymphocytes/metabolism , Rats, Inbred Dahl , Kidney/metabolism , Hypertension/metabolism , Blood Pressure , Sodium Chloride, Dietary/metabolism , Albumins/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 325(4): R411-R422, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37519252

ABSTRACT

Renal denervation (RDN) is a potential therapy for drug-resistant hypertension. However, whether its effects are mediated by ablation of efferent or afferent renal nerves is not clear. Previous studies have implicated that renal inflammation and the sympathetic nervous system are driven by the activation of afferent and efferent renal nerves. RDN attenuated the renal inflammation and sympathetic activity in some animal models of hypertension. In the 2 kidney,1 clip (2K1C) model of renovascular hypertension, RDN also decreased sympathetic activity; however, mechanisms underlying renal and central inflammation are still unclear. We tested the hypothesis that the mechanisms by which total RDN (TRDN; efferent + afferent) and afferent-specific RDN (ARDN) reduce arterial pressure in 2K1C rats are the same. Male Sprague-Dawley rats were instrumented with telemeters to measure mean arterial pressure (MAP), and after 7 days, a clip was placed on the left renal artery. Rats underwent TRDN, ARDN, or sham surgery of the clipped kidney and MAP was measured for 6 wk. Weekly measurements of water intake (WI), urine output (UO), and urinary copeptin were conducted, and urine was analyzed for cytokines/chemokines. Neurogenic pressor activity (NPA) was assessed at the end of the protocol calculated by the depressor response after intraperitoneal injection of hexamethonium. Rats were euthanized and the hypothalamus and kidneys removed for measurement of cytokine content. MAP, NPA, WI, and urinary copeptin were significantly increased in 2K1C-sham rats, and these responses were abolished by both TRDN and ARDN. 2K1C-sham rats presented with renal and hypothalamic inflammation and these responses were largely mitigated by TRDN and ARDN. We conclude that RDN attenuates 2K1C hypertension primarily by ablation of afferent renal nerves which disrupts bidirectional renal neural-immune pathways.NEW & NOTEWORTHY Hypertension resulting from reduced perfusion of the kidney is dependent on renal sensory nerves, which are linked to inflammation in the kidney and hypothalamus. Afferent renal nerves are required for chronic increases in both water intake and vasopressin release observed following renal artery stenosis. Findings from this study suggest an important role of renal sensory nerves that has previously been underestimated in the pathogenesis of 2K1C hypertension.


Subject(s)
Hypertension, Renovascular , Hypertension , Nephritis , Rats , Male , Animals , Rats, Sprague-Dawley , Kidney , Sympathetic Nervous System , Hypothalamus , Inflammation , Blood Pressure/physiology
4.
Am J Physiol Renal Physiol ; 323(3): F349-F360, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35900340

ABSTRACT

Aromatase is a monooxygenase that catalyzes the rate-limiting step of estrogen biosynthesis from androgens. Aromatase inhibitors are widely used for the treatment of patients with hormone receptor-positive breast cancer. However, the effects of aromatase inhibitors on cardiovascular and renal health in females are understudied. Given that estrogen is protective against cardiovascular and kidney diseases, we hypothesized that aromatase inhibition elevates blood pressure and induces kidney injury in female Sprague-Dawley rats. Twelve-week-old female rats were implanted with radiotelemetry transmitters to continuously monitor blood pressure. After baseline blood pressure recording, rats were randomly assigned to treatment with the aromatase inhibitor anastrozole (ASZ) or vehicle (Veh) in drinking water. Twenty days after treatment initiation, rats were shifted from a normal-salt (NS) diet to a high-salt (HS) diet for an additional 40 days. Rats were euthanized 60 days after treatment initiation. Body weight increased in both groups over the study period, but the increase was greater in the ASZ-treated group than in the Veh-treated group. Mean arterial pressure increased in ASZ-treated rats during the NS and HS diet phases but remained unchanged in Veh-treated rats. In addition, urinary excretion of albumin and kidney injury marker-1 and plasma urea were increased in response to aromatase inhibition. Furthermore, histological assessment revealed that ASZ treatment increased morphological evidence of renal tubular injury and proximal tubular brush border loss. In conclusion, chronic aromatase inhibition in vivo with ASZ increases blood pressure and markers of renal proximal tubular injury in female Sprague-Dawley rats, suggesting an important role for aromatization in the maintenance cardiovascular and renal health in females.NEW & NOTEWORTHY Aromatase enzyme catalyzes the rate-limiting step in estrogen biosynthesis. Aromatase inhibitors are clinically used for the treatment of patients with breast cancer; however, the impact of inhibiting aromatization on blood pressure and renal function is incompletely understood. The present findings demonstrate that systemic anastrozole treatment increases blood pressure and renal tubular injury markers in female rats fed a high-salt diet, suggesting an important role for aromatization in preserving cardiovascular and renal health in females.


Subject(s)
Aromatase Inhibitors , Hypertension , Anastrozole/adverse effects , Animals , Aromatase Inhibitors/adverse effects , Biomarkers , Blood Pressure , Estrogens , Female , Hypertension/chemically induced , Kidney/pathology , Neoplasms , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/adverse effects
6.
Front Physiol ; 13: 1085856, 2022.
Article in English | MEDLINE | ID: mdl-36699690
7.
Hypertension ; 73(3): 630-639, 2019 03.
Article in English | MEDLINE | ID: mdl-30595123

ABSTRACT

mTOR (mammalian target of rapamycin) signaling has emerged as a key regulator in a wide range of cellular processes ranging from cell proliferation, immune responses, and electrolyte homeostasis. mTOR consists of 2 distinct protein complexes, mTORC1 (mTOR complex 1) and mTORC2 (mTOR complex 2) with distinct downstream signaling events. mTORC1 has been implicated in pathological conditions, such as cancer and type 2 diabetes mellitus in humans, and inhibition of this pathway with rapamycin has been shown to attenuate salt-induced hypertension in Dahl salt-sensitive rats. Several studies have found that the mTORC2 pathway is involved in the regulation of renal tubular sodium and potassium transport, but its role in hypertension has remained largely unexplored. In the present study, we, therefore, determined the effect of mTORC2 inhibition with compound PP242 on salt-induced hypertension and renal injury in salt-sensitive rats. We found that PP242 not only completely prevented but also reversed salt-induced hypertension and kidney injury in salt-sensitive rats. PP242 exhibited potent natriuretic actions, and chronic administration tended to produce a negative Na+ balance even during high-salt feeding. The results indicate that mTORC2 and the related downstream associated pathways play an important role in regulation of sodium balance and arterial pressure regulation in salt-sensitive rats. Therapeutic suppression of the mTORC2 pathway represents a novel pathway for the potential treatment of hypertension.


Subject(s)
Acute Kidney Injury/prevention & control , Blood Pressure/drug effects , Hypertension/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Hypertension/chemically induced , Hypertension/metabolism , Immunosuppressive Agents/pharmacology , Male , Rats , Rats, Inbred Dahl , Signal Transduction/drug effects , Sodium Chloride, Dietary/toxicity
8.
Front Physiol ; 9: 848, 2018.
Article in English | MEDLINE | ID: mdl-30038578

ABSTRACT

Salt-sensitive hypertension is common in glucocorticoid excess. Glucocorticoid resistance also presents with hypercortisolemia and hypertension but the relationship between salt intake and blood pressure (BP) is not well defined. GRßgeo/+ mice have global glucocorticoid receptor (GR) haploinsufficiency and increased BP. Here we examined the effect of high salt diet on BP, salt excretion and renal blood flow in GRßgeo/+mice. Basal BP was ∼10 mmHg higher in male GRßgeo/+ mice than in GR+/+ littermates. This modest increase was amplified by ∼10 mmHg following a high-salt diet in GRßgeo/+ mice. High salt reduced urinary aldosterone excretion but increased renal mineralocorticoid receptor expression in both genotypes. Corticosterone, and to a lesser extent deoxycorticosterone, excretion was increased in GRßgeo/+ mice following a high-salt challenge, consistent with enhanced 24 h production. GR+/+ mice increased fractional sodium excretion and reduced renal vascular resistance during the high salt challenge, retaining neutral sodium balance. In contrast, sodium excretion and renal vascular resistance did not adapt to high salt in GRßgeo/+ mice, resulting in transient sodium retention and sustained hypertension. With high-salt diet, Slc12a3 and Scnn1a mRNAs were higher in GRßgeo/+ than controls, and this was reflected in an exaggerated natriuretic response to thiazide and benzamil, inhibitors of NCC and ENaC, respectively. Reduction in GR expression causes salt-sensitivity and an adaptive failure of the renal vasculature and tubule, most likely reflecting sustained mineralocorticoid receptor activation. This provides a mechanistic basis to understand the hypertension associated with loss-of-function polymorphisms in GR in the context of habitually high salt intake.

9.
Physiol Genomics ; 50(6): 440-447, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29602296

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Inflammation/genetics , Kidney Medulla/physiology , Kidney Medulla/physiopathology , Metabolic Networks and Pathways/genetics , Perfusion , Animals , Bayes Theorem , Cell Respiration , Hypertension/genetics , Metabolome , Metabolomics , Mitochondria/metabolism , Rats, Inbred Dahl , Regression Analysis , Software
10.
J Am Heart Assoc ; 6(9)2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28899893

ABSTRACT

BACKGROUND: ATP and derivatives are recognized to be essential agents of paracrine signaling. It was reported that ATP is an important regulator of the pressure-natriuresis mechanism. Information on the sources of ATP, the mechanisms of its release, and its relationship to blood pressure has been limited by the inability to precisely measure dynamic changes in intrarenal ATP levels in vivo. METHODS AND RESULTS: Newly developed amperometric biosensors were used to assess alterations in cortical ATP concentrations in response to changes in renal perfusion pressure (RPP) in anesthetized Sprague-Dawley rats. RPP was monitored via the carotid artery; ligations around the celiac/superior mesenteric arteries and the distal aorta were used for manipulation of RPP. Biosensors were acutely implanted in the renal cortex for assessment of ATP. Rise of RPP activated diuresis/natriuresis processes, which were associated with elevated ATP. The increases in cortical ATP concentrations were in the physiological range (1-3 µmol/L) and would be capable of activating most of the purinergic receptors. There was a linear correlation with every 1-mm Hg rise in RPP resulting in a 70-nmol/L increase in ATP. Furthermore, this elevation of RPP was accompanied by a 2.5-fold increase in urinary H2O2. CONCLUSIONS: Changes in RPP directly correlate with renal sodium excretion and the elevation of cortical ATP. Given the known effects of ATP on regulation of glomerular filtration and tubular transport, the data support a role for ATP release in the rapid natriuretic responses to acute increases in RPP.


Subject(s)
Adenosine Triphosphate/metabolism , Blood Pressure , Kidney/blood supply , Kidney/metabolism , Mechanotransduction, Cellular , Paracrine Communication , Renal Circulation , Animals , Biosensing Techniques , Hydrogen Peroxide/urine , Male , Natriuresis , Rats, Sprague-Dawley , Receptors, Purinergic P2/metabolism , Time Factors
11.
Hypertension ; 70(3): 543-551, 2017 09.
Article in English | MEDLINE | ID: mdl-28696224

ABSTRACT

Renal T-cell infiltration is a key component of salt-sensitive hypertension in Dahl salt-sensitive (SS) rats. Here, we use an electronic servo-control technique to determine the contribution of renal perfusion pressure to T-cell infiltration in the SS rat kidney. An aortic balloon occluder placed around the aorta between the renal arteries was used to maintain perfusion pressure to the left kidney at control levels, ≈128 mm Hg, during 7 days of salt-induced hypertension, whereas the right kidney was exposed to increased renal perfusion pressure that averaged 157±4 mm Hg by day 7 of high-salt diet. The number of infiltrating T cells was compared between the 2 kidneys. Renal T-cell infiltration was significantly blunted in the left servo-controlled kidney compared with the right uncontrolled kidney. The number of CD3+, CD3+CD4+, and CD3+CD8+ T cells were all significantly lower in the left servo-controlled kidney. This effect was not specific to T cells because CD45R+ (B cells) and CD11b/c+ (monocytes and macrophages) cell infiltrations were all exacerbated in the hypertensive kidneys. Increased renal perfusion pressure was also associated with augmented renal injury, with increased protein casts and glomerular damage in the hypertensive kidney. Levels of norepinephrine were comparable between the 2 kidneys, suggestive of equivalent sympathetic innervation. Renal infiltration of T cells was not reversed by the return of renal perfusion pressure to control levels after 7 days of salt-sensitive hypertension. We conclude that increased pressure contributes to the initiation of renal T-cell infiltration during the progression of salt-sensitive hypertension in SS rats.


Subject(s)
Cell Movement/immunology , Hypertension , Kidney , T-Lymphocytes , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Hypertension/etiology , Hypertension/pathology , Hypertension/physiopathology , Kidney/blood supply , Kidney/immunology , Kidney/pathology , Rats , Rats, Inbred Dahl , Renal Artery/physiopathology , Sodium Chloride/pharmacology , T-Lymphocytes/pathology , T-Lymphocytes/physiology
12.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26951843

ABSTRACT

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Craving/physiology , Hypertension/genetics , Mineralocorticoid Excess Syndrome, Apparent/physiopathology , Nerve Tissue Proteins/deficiency , Receptors, Mineralocorticoid/physiology , Sodium Chloride, Dietary/toxicity , Solitary Nucleus/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/physiology , Animals , Baroreflex/drug effects , Corticosterone/blood , Dexamethasone/pharmacology , Drinking Behavior , Genes, Synthetic , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mineralocorticoid Excess Syndrome, Apparent/drug therapy , Mineralocorticoid Excess Syndrome, Apparent/genetics , Mineralocorticoid Receptor Antagonists/therapeutic use , Nephrons/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nestin/genetics , Neurons/physiology , Potassium/urine , RNA, Messenger/biosynthesis , Reflex, Abnormal , Solitary Nucleus/physiopathology , Spironolactone/pharmacology
13.
J Vis Exp ; (104)2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26485400

ABSTRACT

Enzymatic microelectrode biosensors have been widely used to measure extracellular signaling in real-time. Most of their use has been limited to brain slices and neuronal cell cultures. Recently, this technology has been applied to the whole organs. Advances in sensor design have made possible the measuring of cell signaling in blood-perfused in vivo kidneys. The present protocols list the steps needed to measure ATP and H2O2 signaling in the rat kidney interstitium. Two separate sensor designs are used for the ex vivo and in vivo protocols. Both types of sensor are coated with a thin enzymatic biolayer on top of a permselectivity layer to give fast responding, sensitive and selective biosensors. The permselectivity layer protects the signal from the interferents in biological tissue, and the enzymatic layer utilizes the sequential catalytic reaction of glycerol kinase and glycerol-3-phosphate oxidase in the presence of ATP to produce H2O2. The set of sensors used for the ex vivo studies further detected analyte by oxidation of H2O2 on a platinum/iridium (Pt-Ir) wire electrode. The sensors for the in vivo studies are instead based on the reduction of H2O2 on a mediator coated gold electrode designed for blood-perfused tissue. Final concentration changes are detected by real-time amperometry followed by calibration to known concentrations of analyte. Additionally, the specificity of the amperometric signal can be confirmed by the addition of enzymes such as catalase and apyrase that break down H2O2 and ATP correspondingly. These sensors also rely heavily on accurate calibrations before and after each experiment. The following two protocols establish the study of real-time detection of ATP and H2O2 in kidney tissues, and can be further modified to extend the described method for use in other biological preparations or whole organs.


Subject(s)
Adenosine Triphosphate/analysis , Biosensing Techniques/methods , Hydrogen Peroxide/analysis , Kidney/chemistry , Adenosine Triphosphate/metabolism , Animals , Biosensing Techniques/instrumentation , Computer Systems , Glucose Oxidase , Glycerolphosphate Dehydrogenase/chemistry , Gold/chemistry , Hydrogen Peroxide/metabolism , Kidney/metabolism , Microelectrodes , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley
14.
Psychoneuroendocrinology ; 59: 59-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26036451

ABSTRACT

Stress or elevated glucocorticoids during sensitive windows of fetal development increase the risk of neuropsychiatric disorders in adult rodents and humans, a phenomenon known as glucocorticoid programming. 11ß-Hydroxysteroid dehydrogenase type 2 (11ß-HSD2), which catalyses rapid inactivation of glucocorticoids in the placenta, controls access of maternal glucocorticoids to the fetal compartment, placing it in a key position to modulate glucocorticoid programming of behavior. However, the importance of the high expression of 11ß-HSD2 within the midgestational fetal brain is unknown. To examine this, a brain-specific knockout of 11ß-HSD2 (HSD2BKO) was generated and compared to wild-type littermates. HSD2BKO have markedly diminished fetal brain 11ß-HSD2, but intact fetal body and placental 11ß-HSD2 and normal fetal and placental growth. Despite normal fetal plasma corticosterone, HSD2BKO exhibit elevated fetal brain corticosterone levels at midgestation. As adults, HSD2BKO show depressive-like behavior and have cognitive impairments. However, unlike complete feto-placental deficiency, HSD2BKO show no anxiety-like behavioral deficits. The clear mechanistic separation of the programmed components of depression and cognition from anxiety implies distinct mechanisms of pathogenesis, affording potential opportunities for stratified interventions.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Anxiety Disorders/enzymology , Brain/embryology , Brain/enzymology , Depressive Disorder/enzymology , Animals , Disease Models, Animal , Female , Fetal Development , Glucocorticoids/blood , Glucocorticoids/metabolism , Male , Maternal-Fetal Exchange/physiology , Mice , Mice, Knockout , Placenta/metabolism , Pregnancy , Risk Factors , Stress, Physiological
15.
Hypertension ; 65(3): 561-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25489057

ABSTRACT

Null mutations in the p67(phox) subunit of nicotinamide adenine dinucleotide phosphate-oxidase confer protection from salt sensitivity on Dahl salt-sensitive rats. Here, we track the sequential changes in medullary blood flow (MBF), glomerular filtration rate (GFR), urinary protein, and mean arterial pressure in SSp67(phox) null rats and wild-type littermates during 21 days of 4.0% NaCl high-salt (HS) diet. Optical fibers were implanted in the renal medulla and MBF was measured in conscious rats by laser Doppler flowmetry. Separate groups of rats were prepared with femoral venous catheters and GFR was measured by the transcutaneous assessment of fluorescein isothiocyanate-sinistrin disappearance curves. Mean arterial blood pressure was measured by telemetry. In wild-type rats, HS caused a rapid reduction in MBF, which was significantly lower than control values by HS day-6. Reduced MBF was associated with a progressive increase in mean arterial pressure, averaging 170±5 mm Hg by HS salt day-21. A significant reduction in GFR was evident on day-14 HS, after the onset of hypertension and reduced MBF. In contrast, HS had no significant effect on MBF in SSp67(phox) null rats and the pressor response to sodium was blunted, averaging 150±3 mm Hg on day-21 HS. GFR was maintained throughout the study and proteinuria was reduced. In summary, when p67(phox) is not functional in the salt-sensitive rats, HS does not cause reduced MBF and salt-sensitive hypertension is attenuated, and consequently renal injury is reduced and GFR is maintained.


Subject(s)
Glomerular Filtration Rate/drug effects , Hypertension/prevention & control , Kidney Medulla/blood supply , Mutation/genetics , NADH, NADPH Oxidoreductases/deficiency , Regional Blood Flow/drug effects , Sodium Chloride, Dietary/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Creatinine/metabolism , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Male , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/physiology , Proteinuria/physiopathology , Proteinuria/prevention & control , Rats , Rats, Inbred Dahl , Rats, Mutant Strains , Reactive Oxygen Species/metabolism , Regional Blood Flow/physiology , Sodium Chloride, Dietary/adverse effects
16.
Purinergic Signal ; 10(1): 71-101, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24265071

ABSTRACT

The involvement of purinergic signalling in kidney physiology and pathophysiology is rapidly gaining recognition and this is a comprehensive review of early and recent publications in the field. Purinergic signalling involvement is described in several important intrarenal regulatory mechanisms, including tuboglomerular feedback, the autoregulatory response of the glomerular and extraglomerular microcirculation and the control of renin release. Furthermore, purinergic signalling influences water and electrolyte transport in all segments of the renal tubule. Reports about purine- and pyrimidine-mediated actions in diseases of the kidney, including polycystic kidney disease, nephritis, diabetes, hypertension and nephrotoxicant injury are covered and possible purinergic therapeutic strategies discussed.


Subject(s)
Health Status , Kidney Diseases/metabolism , Kidney/physiology , Receptors, Purinergic/physiology , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Animals , Biological Transport/physiology , Humans , Hypertension/diagnosis , Hypertension/metabolism , Kidney Diseases/diagnosis
17.
Hypertension ; 60(3): 684-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22777941

ABSTRACT

In vivo, the enzyme 11ß-hydroxysteroid dehydrogenase type 2 influences ligand access to the mineralocorticoid receptor. Ablation of the encoding gene, HSD11B2, causes the hypertensive syndrome of apparent mineralocorticoid excess. Studies in humans and experimental animals have linked reduced 11ß-hydroxysteroid dehydrogenase type 2 activity and salt sensitivity of blood pressure. In the present study, renal mechanisms underpinning salt sensitivity were investigated in Hsd11b2(+/-) mice fed low-, standard-, and high-sodium diets. In wild-type mice, there was a strong correlation between dietary sodium content and fractional sodium excretion but not blood pressure. High sodium feeding abolished amiloride-sensitive sodium reabsorption, consistent with downregulation of the epithelial sodium channel. In Hsd11b2(+/-) mice, the natriuretic response to increased dietary sodium content was blunted, and epithelial sodium channel activity persisted. High-sodium diet also reduced renal blood flow and increased blood pressure in Hsd11b2(+/-) mice. Aldosterone was modulated by dietary sodium in both genotypes, and salt sensitivity in Hsd11b2(+/-) mice was associated with increased plasma corticosterone levels. Chronic administration of an epithelial sodium channel blocker or a glucocorticoid receptor antagonist prevented salt sensitivity in Hsd11b2(+/-) mice, whereas mineralocorticoid receptor blockade with spironolactone did not. This study shows that reduced 11ß-hydroxysteroid dehydrogenase type 2 causes salt sensitivity of blood pressure because of impaired renal natriuretic capacity. This reflects deregulation of epithelial sodium channels and increased renal vascular resistance. The phenotype is not caused by illicit activation of mineralocorticoid receptors by glucocorticoids but by direct activation of glucocorticoid receptors.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Down-Regulation/physiology , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/physiology , Heterozygote , Salt Tolerance/genetics , Salt Tolerance/physiology , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Corticosterone/blood , Genotype , Kidney/blood supply , Mice , Mice, Inbred Strains , Models, Animal , Phenotype , Receptors, Glucocorticoid/physiology , Regional Blood Flow/drug effects , Sodium, Dietary/pharmacology , Vascular Resistance/physiology
18.
Am J Physiol Renal Physiol ; 303(4): F494-502, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22622456

ABSTRACT

In aldosterone target tissues, 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) is coexpressed with mineralocorticoid receptors (MR) and protects the receptor from activation by glucocorticoids. Null mutations in the encoding gene, HSD11B2, cause apparent mineralocorticoid excess, in which hypertension is thought to reflect volume expansion secondary to sodium retention. Hsd11b2(-/-) mice are indeed hypertensive, but impaired natriuretic capacity is associated with significant volume contraction, suggestive of a urine concentrating defect. Water turnover and the urine concentrating response to a 24-h water deprivation challenge were therefore assessed in Hsd11b2(-/-) mice and controls. Hsd11b2(-/-) mice have a severe and progressive polyuric/polydipsic phenotype. In younger mice (∼2 mo of age), polyuria was associated with decreased abundance of aqp2 and aqp3 mRNA. The expression of other genes involved in water transport (aqp4, slc14a2, and slc12a2) was not changed. The kidney was structurally normal, and the concentrating response to water deprivation was intact. In older Hsd11b2(-/-) mice (>6 mo), polyuria was associated with a severe atrophy of the renal medulla and downregulation of aqp2, aqp3, aqp4, slc14a2, and slc12a2. The concentrating response to water deprivation was impaired, and the natriuretic effect of the loop diuretic bumetanide was lost. In older Hsd11b2(-/-) mice, the V2 receptor agonist desmopressin did not restore full urine concentrating capacity. We find that Hsd11b2(-/-) mice develop nephrogenic diabetes insipidus. Gross changes to renal structure are observed, but these were probably secondary to sustained polyuria, rather than of developmental origin.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Diabetes Insipidus/enzymology , Kidney Concentrating Ability/physiology , Kidney/physiology , Aging , Animals , Diabetes Insipidus/genetics , Gene Expression Regulation , Homeostasis , Kidney/anatomy & histology , Kidney Concentrating Ability/genetics , Mice , Mice, Knockout , Osmolar Concentration , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Water/metabolism , Weight Loss
19.
Physiol Genomics ; 40(3): 158-66, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19920212

ABSTRACT

We investigated the effects on urinary steroid and electrolyte excretion and renal gene expression of chronic infusions of ACTH in the mouse. ACTH caused a sustained increase in corticosteroid excretion; aldosterone excretion was only transiently elevated. There was an increase in the excretion of deoxycorticosterone, a weak mineralocorticoid, to levels of physiological significance. Nevertheless, we observed neither antinatriuresis nor kaliuresis in ACTH-treated mice, and plasma renin activity was not suppressed. We identified no changes in expression of mineralocorticoid target genes. Water turnover was increased in chronic ACTH-treated mice, as were hematocrit and hypertonicity: volume contraction is consistent with high levels of glucocorticoid. ACTH-treated mice exhibited other signs of glucocorticoid excess, such as enhanced weight gain and involution of the thymus. We identified novel ACTH-induced changes in 1) genes involved in vitamin D (Cyp27b1, Cyp24a1, Gc) and calcium (Sgk, Calb1, Trpv5) metabolism associated with calciuria and phosphaturia; 2) genes that would be predicted to desensitize the kidney to glucocorticoid action (Nr3c1, Hsd11b1, Fkbp5); and 3) genes encoding transporters of enzyme systems associated with xenobiotic metabolism and oxidative stress. Although there is evidence that ACTH-induced hypertension is a function of physiological cross talk between glucocorticoids and mineralocorticoids, the present study suggests that the major changes in electrolyte and fluid homeostasis and renal function are attributable to glucocorticoids. The calcium and organic anion metabolism pathways that are affected by ACTH may explain some of the known adverse effects associated with glucocorticoid excess.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Hormones/pharmacology , Kidney/drug effects , Kidney/physiology , Transcription, Genetic , Adrenocorticotropic Hormone/metabolism , Animals , Calcium/metabolism , Gene Expression , Hormones/metabolism , Male , Mice , Mice, Inbred C57BL , Potassium/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...