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










Publication year range
1.
Am J Hypertens ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780971

ABSTRACT

BACKGROUND: High salt (HS) intake induces an augmented hypertensive response to nitric oxide (NO) inhibition, though it causes minimal changes in blood pressure (BP) in NO intact condition. The cause of such augmentation is not known. HS induces tumor necrosis factor-alpha (TNFα) production that causes natriuresis via activation of its' receptor type 1 (TNFR1). We hypothesized that NO deficiency reduces renal TNFR1 activity, leading to enhanced sodium retention and hypertension. METHODS: We examined the changes in renal TNFR1 protein expression (Immunohistochemistry analyses) after HS (4% NaCl) intake in wild-type mice (WT, C57BL6) treated with a NO synthase (NOS) inhibitor, nitro-L-arginine methyl ester (L-NAME; 0.05 mg/min/g; osmotic mini-pump), as well as in endothelial NOS knockout mice (eNOSKO) and compared the responses in WT mice with normal salt (NS; 0.3% NaCl) intake. BP was measured with tail-cuff plethysmography and 24-hour urine collections were made using metabolic cages. RESULTS: HS alone did not alter mean BP in untreated mice (76±3 to 77±1 mmHg) but induced an augmented response in L-NAME treated (106±1 vs 97±2 mmHg) and in eNOSKO (107±2 vs 89±3 mmHg) mice. The percentage area of TNFR1 expression in renal tissue was higher in WT+HS (4.1 + 0.5%) than in WT+NS mice (2.7±0.6%). However, TNFR1 expression was significantly lower in L-NAME treated WT+NS (0.9±0.1%) and in eNOSKO+NS (1.4±0.2%) than in both WT+NS and WT+HS mice. CONCLUSION: These data indicate that TNFR1 activity is downregulated in NO deficient conditions, which facilitates salt retention leading to augmented hypertension during HS intake.

2.
3.
Brain Inj ; 37(4): 303-307, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36519359

ABSTRACT

Second impact syndrome (SIS) is an uncommon, but devastating sports-related structural brain injury that results from a second head injury before complete recovery from an initial concussion. The pathophysiology of second impact syndrome is poorly understood, but is hypothesized to involve loss of autoregulation, diffuse cerebral edema, with progression to rapid brain herniation syndromes. Here, we present a case of second impact syndrome in an adolescent high school football player who experienced acute brain herniation and coma. Following stabilization, the patient underwent comprehensive, multidisciplinary rehabilitation in order to achieve significant recovery. A narrative detailing the patient's recovery from one-year post-injury is reviewed.


Subject(s)
Athletic Injuries , Brain Concussion , Football , Adolescent , Humans , Syndrome , Brain Concussion/complications , Brain Concussion/diagnostic imaging , Athletic Injuries/complications , Football/injuries , Athletes , Continuity of Patient Care
4.
Allergy ; 77(4): 1165-1179, 2022 04.
Article in English | MEDLINE | ID: mdl-34800294

ABSTRACT

BACKGROUND: IL-5-dependent residential and IL-18-transformed pathogenic eosinophils have been reported; however, the role of IL-18-transformed CD274-expressing pathogenic eosinophils compared to IL-5-generated eosinophils in promoting airway obstruction in asthma has not yet been examined. METHODS: Eosinophils are detected by tissue anti-MBP and anti-EPX immunostaining, CD274 expression by flow cytometry, and airway resistance using the Buxco FinePointe RC system. RESULTS: We show that A. fumigatus-challenged wild-type mice, and different gene-deficient mice including naïve CC10-IL-18-transgenic mice, accumulate mostly peribronchial and perivascular CD274-expressing eosinophils except naïve CD2-IL-5-transgenic mice. Additionally, we show that CD2-IL-5 transgenic mice following rIL-18 treatment accumulate high number of CD274-expressing perivascular and peribronchial eosinophils with induced collagen, goblet cell hyperplasia and airway resistance compared to saline-challenged CD2-IL5 transgenic mice. Furthermore, we also show that even A. fumigatus-challenged IL-5 -/- mice and rIL-18 given ΔdblGATA mice accumulate CD274-expressing eosinophil-associated asthma pathogenesis including airway obstruction. Most importantly, we provide evidence that neutralization of CD274 and IL-18 in A. fumigatus-challenged mice ameliorate experimental asthma. Taken together, the data presented are clinically significant in establishing that anti-IL-18 neutralization is a novel immunotherapy to restrict asthma pathogenesis. CONCLUSIONS: We demonstrate that IL-18 is critical for inducing asthma pathogenesis, and neutralization of CD274 is a potential immunotherapeutic strategy for asthma.


Subject(s)
Airway Obstruction , Asthma , Airway Obstruction/etiology , Airway Obstruction/pathology , Animals , Asthma/metabolism , B7-H1 Antigen/metabolism , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Eosinophils/metabolism , Humans , Interleukin-18/metabolism , Interleukin-5/metabolism , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic
5.
Physiol Rep ; 9(16): e14990, 2021 08.
Article in English | MEDLINE | ID: mdl-34427402

ABSTRACT

In hypertension induced by angiotensin II (AngII) administration with high salt (HS) intake, intrarenal angiotensinogen (AGT) and tumor necrosis factor-alpha (TNF-α) levels increase. However, TNF-α has been shown to suppress AGT formation in cultured renal proximal tubular cells. We examined the hypothesis that elevated AngII levels during HS intake reduces TNF-α receptor type 1 (TNFR1) activity in the kidneys, thus facilitating increased intrarenal AGT formation. The responses to HS diet (4% NaCl) with chronic infusion of AngII (25 ng/min) via implanted minipump for 4 weeks were assessed in wild-type (WT) and knockout (KO) mice lacking TNFR1 or TNFR2 receptors. Blood pressure was measured by tail-cuff plethysmography, and 24-h urine samples were collected using metabolic cages prior to start (0 day) and at the end of 2nd and 4th week periods. The urinary excretion rate of AGT (uAGT; marker for intrarenal AGT) was measured using ELISA. HS +AngII treatment for 4 weeks increased mean arterial pressure (MAP) in all strains of mice. However, the increase in MAP in TNFR1KO (77 ± 2 to 115 ± 3 mmHg; n = 7) was significantly greater (p < 0.01) than in WT (76 ± 1 to 102 ± 2 mmHg; n = 7) or in TNFR2KO (78 ± 2 to 99 ± 5 mmHg; n = 6). The increase in uAGT at 4th week was also greater (p < 0.05) in TNFR1KO mice (6 ± 2 to 167 ± 75 ng/24 h) than that in WT (6 ± 3 to 46 ± 16 ng/24 h) or in TNFR2KO mice (8 ± 7 to 65 ± 44 ng/24 h). The results indicate that TNFR1 exerts a protective role by mitigating intrarenal AGT formation induced by elevated AngII and HS intake.


Subject(s)
Angiotensinogen/metabolism , Hypertension, Renal/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Angiotensin II/toxicity , Animals , Blood Pressure , Hypertension, Renal/etiology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Sodium Chloride, Dietary/toxicity
6.
Physiol Rep ; 9(15): e14942, 2021 08.
Article in English | MEDLINE | ID: mdl-34337896

ABSTRACT

Intravenous infusion of relatively higher doses of angiotensin II (AngII) elicits natriuresis as opposed to its usual anti-natruretic response. As AngII can induce tumor necrosis factor-α (TNFα) production which elicits natriuresis via its action on TNFα receptor type 1 (TNFR1), we hypothesize that the concomitant release of TNFα contributes to the natriuretic response to AngII. Responses to AngII infusion (1 ng min-1  g-1 for 75 min, iv) were evaluated in anesthetized knockout (KO) mice lacking TNFR1 (n = 6) and TNFR2 (TNFα receptor type 2; n = 6) and compared these responses with those in wild type (WT; n = 6) mice. Arterial pressure (AP) was recorded from a cannula placed in the carotid artery. Renal blood flow (RBF) and glomerular filtration rate (GFR) were measured by PAH and inulin clearances, respectively. Urine was collected from a catheter placed in the bladder. AngII caused similar increases (p < 0.05 vs basal values) in AP (WT, 37 ± 5%; TNFR1KO, 35 ± 4%; TNFR2KO, 30 ± 4%) and decreases (p < 0.05) in RBF (WT, -39 ± 5%; TNFR1KO, -28 ± 6%; TNFR2KO, -31 ± 4%) without significant changes in GFR (WT, -17 ± 7%; TNFR1KO, -18 ± 7%; TNFR2KO, -12 ± 7%). However, despite similar changes in AP and renal hemodynamics, AngII induced increases (p < 0.05) in urinary sodium excretion in WT (3916 ± 942%) were less in the KO strains, more or less in TNFR1KO (473 ± 170%) than in TNFR2KO (1176 ± 168%). These data indicate that TNF-α receptors, particularly TNFR1 are involved in the natriuretic response that occur during acute infusion of AngII and thus, plays a protective role in preventing excessive salt retention at clinical conditions associated with elevated AngII level.


Subject(s)
Angiotensin II/toxicity , Kidney Diseases/prevention & control , Natriuresis/drug effects , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Sodium/metabolism , Animals , Blood Pressure , Glomerular Filtration Rate , Hemodynamics , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Renal Circulation
7.
Physiol Rep ; 8(24): e14621, 2021 01.
Article in English | MEDLINE | ID: mdl-33345460

ABSTRACT

High salt (HS) intake is usually considered as an aggravating factor to induce inflammatory renal injury. However, the changes in the renal levels of inflammatory cytokines during HS intake is not yet clearly defined. We hypothesize that HS increases renal levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) but decreases interleukin-10 (IL-10; anti-inflammatory cytokine) and these responses exacerbate in NO deficient conditions. Both wild-type (WT) and endothelial NO synthase knockout (eNOSKO) mice (~8 weeks old, n = 6 in each group) were given normal-salt (NS; 0.3% NaCl) and HS (4% NaCl) containing diets for 2 weeks. Systolic blood pressure (SBP) was determined by tail-cuff plethysmography and urine collections were made using metabolic cages. Basal SBP was higher in eNOSKO than WT mice (131 ± 7 vs 117 ± 3 mmHg; p < .05). HS intake for 2 weeks increased SBP in eNOSKO (161 ± 5 mmHg) but not in WT mice. In NS groups, the cytokine levels in renal tissues (measured using ELISA kits and expressed in pg/mg protein) were significantly higher in eNOSKO than WT mice (TNF-α, 624 ± 67 vs. 325 ± 73; IL-6, 619 ± 106 vs. 166 ± 61; IL-10, 6,087 ± 567 vs. 3,929 ± 378). Interestingly, these cytokine levels in HS groups were significantly less both in WT (TNF-α, 114 ± 17; IL-6, 81 ± 14; IL-10, 865 ± 130) and eNOSKO (TNF-α, 115 ± 18; IL-6, 56 ± 7; IL-10, 882 ± 141) mice. These findings indicate that HS induces downregulation of cytokines in the kidney. Such HS-induced reduction in cytokines, particularly TNF-α (a natriuretic agent), would facilitate more salt-retention, and thus, leading to salt-sensitive hypertension in NO deficient conditions.


Subject(s)
Interleukin-10/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Sodium Chloride, Dietary/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood Pressure , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics
8.
Front Physiol ; 11: 559341, 2020.
Article in English | MEDLINE | ID: mdl-33281610

ABSTRACT

In the kidney, the stimulation of renin production by the collecting duct (CD-renin) contributes to the development of hypertension. The CD is a major nephron segment for the synthesis of nitric oxide (NO), and low NO bioavailability in the renal medulla is associated with hypertension. However, it is unknown whether NO regulates renin production in the CD. To test the hypothesis that low intrarenal NO levels stimulate the production of CD-renin, we first examined renin expression in the distal nephron segments of CD-eNOS deficient mice. In these mice, specific CD-renin immunoreactivity was increased compared to wild-type littermates; however, juxtaglomerular (JG) renin was not altered. To further assess the intracellular mechanisms involved, we then treated M-1 cells with either 1 mM L-NAME (L-arginine analog), an inhibitor of NO synthase activity, or 1 mM NONOate, a NO donor. Both treatments increased intracellular renin protein levels in M-1 cells. However, only the inhibition of NOS with L-NAME stimulated renin synthesis and secretion as reflected by the increase in Ren1C transcript and renin protein levels in the extracellular media, respectively. In addition, NONOate induced a fast mobilization of cGMP and intracellular renin accumulation. These response was partially prevented by guanylyl cyclase inhibition with ODQ (1H-[1,2,4] oxadiazolo[4,3-a]quinoxalin-1]. Accumulation of intracellular renin was blocked by protein kinase G (PKG) and protein kinase C (PKC) inhibitors. Our data indicate that low NO bioavailability increases CD-renin synthesis and secretion, which may contribute to the activation of intrarenal renin angiotensin system.

9.
Hypertension ; 70(4): 839-845, 2017 10.
Article in English | MEDLINE | ID: mdl-28847894

ABSTRACT

IL-10 (interleukin-10) has been suggested to play a protective role in angiotensin II (AngII)-induced cardiovascular disorders. This study examined the role of endogenous IL-10 in salt-sensitive hypertension and renal injury induced by AngII. Responses to chronic AngII (400 ng/min per kilogram body weight; osmotic minipump) infusion were evaluated in IL-10 gene knockout mice fed with either normal salt diet (0.3% NaCl) or high salt (HS; 4% NaCl) diet, and these responses were compared with those in wild-type mice. Normal salt diets or HS diets were given alone for the first 2 weeks and then with AngII treatment for an additional 2 weeks (n=6 in each group). Arterial pressure was continuously monitored by implanted radio-telemetry, and a 24-hour urine collection was performed by metabolic cages on the last day of the experimental period. Basal mean arterial pressure was lower in IL-10 gene knockout mice than in wild-type (98±3 versus 113±3 mm Hg) mice. Mean arterial pressure responses to normal salt/HS alone or to the AngII+normal salt treatment were similar in both strains. However, the increase in mean arterial pressure induced by the AngII+HS treatment was significantly lower in IL-10 gene knockout mice (15±5% versus 37±3%) compared with wild-type mice. Renal tissue endothelial nitric oxide synthase expression (≈3-folds) and urinary excretion of nitric oxide metabolites, nitrate/nitrite (1.2±0.1 versus 0.2±0.02 µmol/L/24 hours) were higher in IL-10 gene knockout mice compared with wild-type mice. These results indicate that an increase in nitric oxide production helps to mitigate salt-sensitive hypertension induced by AngII and suggest that a compensatory interaction between IL-10 and nitric oxide exists in modulating AngII-induced responses during HS intake.


Subject(s)
Angiotensin II/metabolism , Blood Pressure , Hypertension , Interleukin-10/metabolism , Kidney , Sodium Chloride, Dietary , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Sodium Chloride, Dietary/metabolism , Sodium Chloride, Dietary/pharmacology
10.
Am J Physiol Renal Physiol ; 313(4): F1005-F1008, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28724611

ABSTRACT

Hypertension is considered to be a low-grade inflammatory condition characterized by the presence of various proinflammatory cytokines. Tumor necrosis factor-α (TNF-α) is a constituent of the proinflammatory cytokines that is associated with salt-sensitive hypertension (SSH) and related renal injury. Elevated angiotensin II (ANG II) and other factors such as oxidative stress conditions promote TNF-α formation. Many recent studies have provided evidence that TNF-α exerts a direct renal action by regulating hemodynamic and excretory function in the kidney. The cytokine incites a strong natriuretic response and plays a part in regulation of the intrarenal renin-angiotensin system. The exact mechanistic role of TNF-α in the development of SSH is as yet poorly understood. While TNF-α antagonism has been shown to attenuate hypertensive responses in many hypertensive animal models, contrasting findings demonstrate that the direct systemic administration of TNF-α usually induces hypotensive as well as natriuretic responses, indicating a counterregulatory role of TNF-α in SSH. Differential activities of two cell surface receptors of TNF-α (receptor type 1 and type 2) may explain the contradictory functions of TNF-α in the setting of hypertension. This short review will evaluate ongoing research studies that investigate the action of TNF-α within the kidney and its role as an influential pathophysiological variable in the development of SSH and renal injury. This information may help to develop specific TNF-α receptor targeting as an effective treatment strategy in this clinical condition.


Subject(s)
Blood Pressure , Hypertension/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Kidney/metabolism , Sodium Chloride, Dietary/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Humans , Hypertension/immunology , Hypertension/physiopathology , Inflammation/immunology , Inflammation/physiopathology , Inflammation Mediators/immunology , Kidney/immunology , Kidney/physiopathology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Renin-Angiotensin System , Signal Transduction , Tumor Necrosis Factor-alpha/immunology
11.
Hypertension ; 69(6): 1104-1112, 2017 06.
Article in English | MEDLINE | ID: mdl-28416584

ABSTRACT

Cytochrome P450 1B1 protects against angiotensin II (Ang II)-induced hypertension and associated cardiovascular changes in female mice, most likely via production of 2-methoxyestradiol. This study was conducted to determine whether 2-methoxyestradiol ameliorates Ang II-induced hypertension, renal dysfunction, and end-organ damage in intact Cyp1b1-/-, ovariectomized female, and Cyp1b1+/+ male mice. Ang II or vehicle was infused for 2 weeks and administered concurrently with 2-methoxyestradiol. Mice were placed in metabolic cages on day 12 of Ang II infusion for urine collection for 24 hours. 2-Methoxyestradiol reduced Ang II-induced increases in systolic blood pressure, water consumption, urine output, and proteinuria in intact female Cyp1b1-/- and ovariectomized mice. 2-Methoxyestradiol also reduced Ang II-induced increase in blood pressure, water intake, urine output, and proteinuria in Cyp1b1+/+ male mice. Treatment with 2-methoxyestradiol attenuated Ang II-induced end-organ damage in intact Cyp1b1-/- and ovariectomized Cyp1b1+/+ and Cyp1b1-/- female mice and Cyp1b1+/+ male mice. 2-Methoxyestradiol mitigated Ang II-induced increase in urinary excretion of angiotensinogen in intact Cyp1b1-/- and ovariectomized Cyp1b1+/+ and Cyp1b1-/- female mice but not in Cyp1b1+/+ male mice. The G protein-coupled estrogen receptor 1 antagonist G-15 failed to alter Ang II-induced increases in blood pressure and renal function in Cyp1b1+/+ female mice. These data suggest that 2-methoxyestradiol reduces Ang II-induced hypertension and associated end-organ damage in intact Cyp1b1-/-, ovariectomized Cyp1b1+/+ and Cyp1b1-/- female mice, and Cyp1b1+/+ male mice independent of G protein-coupled estrogen receptor 1. Therefore, 2-methoxyestradiol could serve as a therapeutic agent for treating hypertension and associated pathogenesis in postmenopausal females, and in males.


Subject(s)
Angiotensin II/pharmacology , Cytochrome P-450 CYP1B1/drug effects , Estradiol/analogs & derivatives , Hypertension/drug therapy , Kidney Diseases/drug therapy , Reactive Oxygen Species/metabolism , 2-Methoxyestradiol , Animals , Blood Pressure/drug effects , Blood Pressure Determination , Cytochrome P-450 CYP1B1/metabolism , Disease Models, Animal , Estradiol/pharmacology , Female , Hypertension/chemically induced , Kidney Diseases/pathology , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovariectomy/methods , Random Allocation , Sensitivity and Specificity , Sex Factors
12.
Physiol Rep ; 4(7)2016 Apr.
Article in English | MEDLINE | ID: mdl-27081160

ABSTRACT

In the normal condition, endogenous formation of peroxynitrite (ONOO-) from the interaction of nitric oxide and superoxide has been suggested to play a renoprotective role. However, the exact mechanism associated with renoprotection by this radical compound is not yet clearly defined. AlthoughONOO- usually inhibits renal tubular Na(+)K(+)ATPase (NKA) activity at high concentrations (micromolar to millimolar range [µM-mM], achieved in pathophysiological conditions), the effects at lower concentrations (nanomolar range [nM], relevant in normal condition) remain unknown. To examine the direct effect ofONOO- onNKAactivity, preparations of cellular membrane fraction from mouse renal tissue and from culturedHK2 cells (human proximal tubular epithelial cell lines) were incubated for 10 and 30 min each with different concentrations ofONOO- (10 nmol/L-200 µmol/L).NKAactivity in these samples (n = 5 in each case) was measured via a colorimetric assay capable of detecting inorganic phosphate. At high concentrations (1-200 µmol/L),ONOO- caused dose-dependent inhibition ofNKAactivity (-3.0 ± 0.6% and -36.4 ± 1.4%). However,NKAactivity remained unchanged at 100 and 500 nmol/LONOO- concentration, but interestingly, at lower concentrations (10 and 50 nmol/L),ONOO- caused small but significant increases in theNKAactivity (3.3 ± 1.1% and 3.1 ± 0.6%). Pretreatment with aONOO- scavenger, mercaptoethylguanidine (MEG; 200 µmol/L), prevented these biphasic responses toONOO-. This dose-dependent biphasic action ofONOO(-)onNKAactivity may implicate that this radical compound helps to maintain sodium homeostasis either by enhancing tubular sodium reabsorption under normal conditions or by inhibiting it during oxidative stress conditions.


Subject(s)
Cell Membrane/drug effects , Enzyme Activators/pharmacology , Epithelial Cells/drug effects , Kidney Tubules, Proximal/drug effects , Peroxynitrous Acid/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Line , Cell Membrane/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Free Radical Scavengers/pharmacology , Humans , Kidney Tubules, Proximal/enzymology , Mice, Inbred C57BL , Nitric Oxide/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Time Factors
13.
Hypertension ; 67(5): 916-26, 2016 May.
Article in English | MEDLINE | ID: mdl-26928804

ABSTRACT

6ß-Hydroxytestosterone, a cytochrome P450 1B1-derived metabolite of testosterone, contributes to the development of angiotensin II-induced hypertension and associated cardiovascular pathophysiology. In view of the critical role of angiotensin II in the maintenance of renal homeostasis, development of hypertension, and end-organ damage, this study was conducted to determine the contribution of 6ß-hydroxytestosterone to angiotensin II actions on water consumption and renal function in male Cyp1b1(+/+) and Cyp1b1(-/-) mice. Castration of Cyp1b1(+/+) mice or Cyp1b1(-/-) gene disruption minimized the angiotensin II-induced increase in water consumption, urine output, proteinuria, and sodium excretion and decreases in urine osmolality. 6ß-Hydroxytestosterone did not alter angiotensin II-induced increases in water intake, urine output, proteinuria, and sodium excretion or decreases in osmolality in Cyp1b1(+/+) mice, but restored these effects of angiotensin II in Cyp1b1(-/-) or castrated Cyp1b1(+/+) mice. Cyp1b1 gene disruption or castration prevented angiotensin II-induced renal fibrosis, oxidative stress, inflammation, urinary excretion of angiotensinogen, expression of angiotensin II type 1 receptor, and angiotensin-converting enzyme. 6ß-Hydroxytestosterone did not alter angiotensin II-induced renal fibrosis, inflammation, oxidative stress, urinary excretion of angiotensinogen, expression of angiotensin II type 1 receptor, or angiotensin-converting enzyme in Cyp1b1(+/+)mice. However, in Cyp1b1(-/-) or castrated Cyp1b1(+/+) mice, it restored these effects of angiotensin II. These data indicate that 6ß-hydroxytestosterone contributes to increased thirst, impairment of renal function, and end-organ injury associated with angiotensin II-induced hypertension in male mice and that cytochrome P450 1B1 could serve as a novel target for treating renal disease and hypertension in male mice.


Subject(s)
Angiotensin II/pharmacology , Cytochrome P-450 CYP1B1/genetics , Hydroxytestosterones/pharmacology , Kidney Diseases/metabolism , Oxidative Stress/drug effects , Analysis of Variance , Animals , Biopsy, Needle , Castration , Disease Models, Animal , Hypertension/physiopathology , Immunohistochemistry , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Function Tests , Male , Mice , RNA, Messenger/analysis , Random Allocation , Reactive Oxygen Species/metabolism , Reference Values , Risk Factors
14.
Curr Hypertens Rev ; 11(1): 38-48, 2015.
Article in English | MEDLINE | ID: mdl-26028244

ABSTRACT

Salt sensitive hypertension is characterized by increases in blood pressure in response to increases in dietary salt intake and is associated with an enhanced risk of cardiovascular and renal morbidity. Although researchers have sought for decades to understand how salt sensitivity develops in humans, the mechanisms responsible for the increases in blood pressure in response to high salt intake are complex and only partially understood. Until now, scientists have been unable to explain why some individuals are salt sensitive and others are salt resistant. Although a central role for the kidneys in the development of salt sensitivity and hypertension has been generally accepted, it is also recognized that hypertension is of multifactorial origin and a variety of factors can induce, or prevent, blood pressure responsiveness to the manipulation of salt intake. Excess salt intake in susceptible persons may also induce inappropriate central and sympathetic nervous system responses and increase the production of intrarenal angiotensin II, catecholamines and other factors such as oxidative stress and inflammatory cytokines. One key factor is the concomitant inappropriate or paradoxical activation of the intrarenal renin-angiotensin system, by high salt intake. This is reflected by the increases in urinary angiotensinogen during high salt intake in salt sensitive models. A complex interaction between neuroendocrine factors and the kidney may underlie the propensity for some individuals to retain salt and develop salt-dependent hypertension. In this review, we focus mainly on the renal contributions that provide the mechanistic links between chronic salt intake and the development of hypertension.


Subject(s)
Angiotensins/physiology , Blood Pressure/drug effects , Hypertension/physiopathology , Kidney/physiopathology , Sodium, Dietary/pharmacology , Angiotensin II/physiology , Angiotensin II/urine , Angiotensins/urine , Animals , Humans , Kidney/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiology , Models, Biological , Oxidative Stress/physiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
15.
Pflugers Arch ; 467(4): 833-41, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24953240

ABSTRACT

While it is clearly recognized that increased intrarenal nitric oxide (NO) levels elicit natriuresis, confounding data showing that systemic nitric oxide synthase inhibition (NOSi) also increases sodium excretion (UNaV) poses a conundrum. This response has been attributed to the associated increases in arterial pressure (AP); however, the increases in AP and in UNaV are temporally dissociated. The changes in regional renal haemodynamics induced by NOSi could also contribute to the alterations of UNaV. To evaluate the roles of AP and non-AP mechanisms mediating the natriuresis, N ω-nitro-L-arginine methyl ester hydrochloride (L-NAME) was infused i.v. at doses ranging from 5 to 50 µg/kg/min in anaesthetized rats. UNaV, perfusion of the cortex (cortical blood flow, CBF) and medulla (medullary blood flow, MBF) with laser-Doppler flowmetry and glomerular filtration rate (GFR) were measured. UNaV increased from 0.6 ± 0.2 to 1.6 ± 0.1 µmol/kg/min (P < 0.05) with the lower nonpressor doses. With the higher doses, AP increased from 116 ± 4 to 122 ± 4 mmHg and UNaV increased from 1.1 ± 0.3 to 3.3 ± 0.7 µmol/min/g (P < 0.002). UNaV increased similarly in a group where renal AP was maintained at baseline levels. The associated reductions in CBF (17 ± 5 and 38 ± 5 %) and MBF (27 ± 6 and 52 ± 6 %) would be expected to attenuate rather than contribute to the natriuresis. Plasma atrial natriuretic peptide (ANP) concentrations increased significantly following NOSi. Anantin, a natriuretic peptide receptor-A blocker, prevented or reversed the L-NAME-induced natriuresis without altering the L-NAME-induced changes in AP or CBF. The results indicate that increased ANP and related natriuretic peptides mediate the AP-independent natriuresis, at least partly, elicited by systemic L-NAME infusion and help resolve the conundrum of natriuresis during systemic NOSi.


Subject(s)
Atrial Natriuretic Factor/blood , Blood Pressure , Natriuresis , Nitric Oxide/metabolism , Animals , Hemodynamics , Kidney/drug effects , Kidney/metabolism , Kidney/physiology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Rats , Rats, Sprague-Dawley , Sodium/metabolism
16.
Physiol Rep ; 2(2): e00228, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24744897

ABSTRACT

Earlier, we demonstrated that the inhibition of nitric oxide synthase (NOS) by nitro-l-arginine methyl ester (l-NAME) infusion increases the endogenous production of proinflammatory cytokine, tumor necrosis factor (TNF-α). In the present study, we examined the hypothesis that inhibition of nitric oxide (NO) production leads to the suppression of interleukin (IL)-10 (anti-inflammatory cytokine) generation which facilitates the enhancement of TNF-α production endogenously. Using appropriate enzyme-linked immunosorbent assay kits and immunohistochemical staining, the levels of IL-10 and TNF-α in plasma (P) and in renal tissues (R) were measured in anesthetized mice (C57BL/6; ~10 weeks age; n = 6/group) infused with or without l-NAME (200 µg/min/kg; i.v. for 2 h). Compared to vehicle-treated control mice, l-NAME-treated mice had a lower level of IL-10 (P, 0.3 ± 0.1 vs. 2.6 ± 0.6 ng/mL; R, 0.5 ± 0.1 vs. 3 ± 0.1 ng/mg protein) and a higher level of TNF-α (P, 432 ± 82 vs. undetected pg/mL; R, 58 ± 7 vs. 6 ± 5 pg/mg protein). IL-10 protein expression, present mostly in the distal nephron segments in control mice, was markedly downregulated in l-NAME-treated mice. Compared to control mice, TNF-α expression increased 2.5-fold in renal cortical sections (mostly in the distal nephron segments) in l-NAME-treated mice. Coinfusion of a NO donor, S-nitroso-N-acetyl-penicillamine (SNAP; 25 µg/min/kg) with l-NAME in a separate group of mice prevented these changes in IL-10 and TNF-α induced by l-NAME. IL-10 infusion (0.075 ng/min/g) in l-NAME-treated mice markedly attenuated l-NAME-induced increments in TNF-α. Thus, these results demonstrate that NOS inhibition decreases endogenous IL-10 generation and thus, minimizes its immune downregulating action on the TNF-α production in the kidney.

17.
Am J Physiol Renal Physiol ; 305(7): F1031-41, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23926180

ABSTRACT

In the present study, we examine the hypothesis that the nitric oxide (NO) produced by endothelial NO synthase (eNOS) plays a protective role in the development of ANG II-induced hypertension and renal injury by minimizing oxidative stress and the inflammation induced by TNF-α. Systolic blood pressure (SBP) and renal injury responses to chronic infusions of ANG II (via implanted minipumps) were evaluated for 2 wk in wild-type (WT) and in eNOS knockout mice (KO) cotreated with or without a superoxide (O2(-)) scavenger, tempol (400 mg/l in the drinking water), or a TNF-α receptor blocker, etanercept (5 mg/kg/day ip). In study 1, when ANG II was given at a dose of 25 ng/min, it increased mean SBP in WT mice (Δ36 ± 3 mmHg; n = 7), and this effect was attenuated in mice pretreated with tempol (Δ24 ± 3 mmHg; n = 6). In KO mice (n = 9), this dose of ANG II resulted in severe renal injury associated with high mortality. To avoid this high mortality in KO, study 2 was conducted with a lower dose of ANG II (10 ng/min) that increased SBP slightly in WT (Δ17 ± 7 mmHg; n = 6) but exaggeratedly in KO (Δ48 ± 12 mmHg, n = 6) associated with severe renal injury. Cotreatment with either tempol (n = 6) or etanercept (n = 6) ameliorated the hypertensive, as well as the renal injury responses in KO compared with WT. These data demonstrate a protective role for eNOS activity in preventing renal inflammatory injury and hypertension induced by chronic increases in ANG II.


Subject(s)
Angiotensin II/physiology , Hypertension/enzymology , Hypertension/prevention & control , Nephritis/enzymology , Nitric Oxide Synthase Type III/physiology , Ribonuclease, Pancreatic/toxicity , Angiogenesis Inducing Agents/toxicity , Angiotensin II/administration & dosage , Animals , Hypertension/etiology , Inflammation/enzymology , Inflammation/pathology , Male , Mice , Mice, Knockout , Nephritis/etiology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Am J Physiol Renal Physiol ; 304(7): F991-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23389459

ABSTRACT

Tumor necrosis factor-alpha (TNF-α) has been implicated in salt-sensitive hypertension and renal injury (RI) induced by angiotensin II (ANG II). To determine the receptor type of TNF-α involved in this mechanism, we evaluated the responses to chronic ANG II infusion (25 ng/min by implanted minipump) given with high-salt diet (HS; 4% NaCl) for 2 wk in gene knockout mice for TNF-α receptor type 1 (TNFR1KO; n = 6) and type 2 (TNFR2KO; n = 6) and compared the responses with those in wild-type (WT; C57BL/6; n = 6) mice. Blood pressure in these mice was measured by implanted radiotelemetry as well as by tail-cuff plethysmography. RI responses were assessed by measuring macrophage cell infiltration (CD68(+) immunohistochemistry), glomerulosclerosis (PAS staining), and interstitial fibrosis (Gomori's trichrome staining) in renal tissues at the end of the treatment period. The increase in mean arterial pressure induced by ANG II + HS treatment was not different in these three groups of mice (TNFR1KO, 114 ± 1 to 161 ± 7 mmHg; TNFR2KO, 113 ± 1 to 161 ± 3 mmHg; WT, 110 ± 3 to 154 ± 3 mmHg). ANG II + HS-induced RI changes were similar in TNFR1KO mice but significantly less in TNFR2KO mice (macrophage infiltration, 0.02 ± 0.01 vs. 1.65 ± 0.45 cells/mm(2); glomerulosclerosis, 26.3 ± 2.6 vs. 35.7 ± 2.2% area; and interstitial fibrosis, 5.2 ± 0.6 vs. 8.1 ± 1.1% area) compared with the RI changes in WT mice. The results suggest that a direct activation of TNF-α receptors may not be required in inducing hypertensive response to chronic ANG II administration with HS intake, but the induction of inflammatory responses leading to renal injury are mainly mediated by TNF-α receptor type 2.


Subject(s)
Angiotensin II/pharmacology , Glomerulonephritis/chemically induced , Receptors, Tumor Necrosis Factor, Type II/metabolism , Sodium Chloride, Dietary/adverse effects , Animals , Blood Pressure/drug effects , Kidney/physiopathology , Kidney Glomerulus/pathology , Macrophages/immunology , Male , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II/genetics , Sodium Chloride, Dietary/administration & dosage , Urination/drug effects , Urination/physiology
19.
Urology ; 81(1): 210.e5-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23153953

ABSTRACT

OBJECTIVE: To investigate the relationship between renal ischemia injury and concentrations of 8-isoprostane in a rat kidney model during renal hilar clamping and their correlation with the administration of allopurinol before clamping. MATERIALS AND METHODS: Reperfusion injury occurs after the reintroduction of blood flow after a prolonged period of ischemia. Thought to be due to oxygen free radicals released by the endothelial, mitochondrial, and parenchymal cells, this process leads to a cascade of events whereby infiltrative leukocytes generate cytokines and reactive oxygen species. The present study was performed in 2 parts. Our primary objective was to first develop a method of quantitating the renal damage using a prostaglandin compound formed in vivo, specifically isoprostane. After the development of this animal model of quantitating renal injury, our second objective was to apply this model and investigate allopurinol's nephroprotective abilities. A microdialysis probe was inserted into the renal parenchyma of rats to allow continuous dialysis and collection of the effluent for isoprostane levels. After clamping of the renal vessels to induce ischemia, the interstitial effluent from the probe was collected and subsequently analyzed for 8-isoprostane levels with and without allopurinol pretreatment. RESULTS: Clamping of the renal hilum in this rat model significantly increased 8-isoprostane levels. After 60 minutes of clamp time, the largest absolute increase in 8-isoprostane levels resulted, representing a 3.2-fold increase from baseline. However, the rats that had been pretreated with allopurinol demonstrated significantly less isoprostane levels, to baseline levels. CONCLUSION: Allopurinol has demonstrated significant benefits by reducing reperfusion injury in rat kidneys, as demonstrated by the use of 8-isoprostane as a tool for the real-time measurement of ischemic injury.


Subject(s)
Allopurinol/therapeutic use , Dinoprost/analogs & derivatives , Free Radical Scavengers/therapeutic use , Kidney/blood supply , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Animals , Dinoprost/metabolism , Disease Models, Animal , Kidney/metabolism , Kidney/pathology , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Renal Artery , Time Factors
20.
Am J Physiol Renal Physiol ; 302(12): F1650-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22461305

ABSTRACT

Acute administration of tumor necrosis factor-α (TNF-α) resulted in decreases in renal blood flow (RBF) and glomerular filtration rate (GFR) but induced diuretic and natriuretic responses in mice. To define the receptor subtypes involved in these renal responses, experiments were conducted to assess the responses to human recombinant TNF-α (0.3 ng·min(-1)·g body wt(-1) iv infusion for 75 min) in gene knockout (KO) mice for TNF-α receptor type 1 (TNFαR1 KO, n = 5) or type 2 (TNFαR2 KO, n = 6), and the results were compared with those obtained in corresponding wild-type [WT (C57BL/6), n = 6] mice. Basal levels of RBF (PAH clearance) and GFR (inulin clearance) were similar in TNFαR1 KO, but were lower in TNFαR2 KO, than WT mice. TNF-α infusion in WT mice decreased RBF and GFR but caused a natriuretic response, as reported previously. In TNFαR1 KO mice, TNF-α infusion failed to cause such vasoconstrictor or natriuretic responses; rather, there was an increase in RBF and a decrease in renal vascular resistance. Similar responses were also observed with infusion of murine recombinant TNF-α in TNFαR1 KO mice (n = 5). However, TNF-α infusion in TNFαR2 KO mice caused changes in renal parameters qualitatively similar to those observed in WT mice. Immunohistochemical analysis in kidney slices from WT mice demonstrated that while both receptor types were generally located in the renal vascular and tubular cells, only TNFαR1 was located in vascular smooth muscle cells. There was an increase in TNFαR1 immunoreactivity in TNFαR2 KO mice, and vice versa, compared with WT mice. Collectively, these functional and immunohistological findings in the present study demonstrate that the activation of TNFαR1, not TNFαR2, is mainly involved in mediating the acute renal vasoconstrictor and natriuretic actions of TNF-α.


Subject(s)
Kidney/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Blood Pressure/drug effects , Glomerular Filtration Rate/drug effects , Kidney/blood supply , Kidney/drug effects , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Renal Circulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...