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

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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.

6.
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
7.
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
8.
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
9.
Am J Physiol Renal Physiol ; 302(1): F85-94, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21900456

ABSTRACT

Augmentation of intrarenal angiotensinogen (AGT) synthesis, secretion, and excretion is associated with the development of hypertension, renal oxidative stress, and tissue injury during ANG II-dependent hypertension. High salt (HS) exacerbates hypertension and kidney injury, but the mechanisms remain unclear. In this study, we determined the consequences of HS intake alone compared with chronic ANG II infusion and combined HS plus ANG II on the stimulation of urinary AGT (uAGT), renal oxidative stress, and renal injury markers. Sprague-Dawley rats were subjected to 1) a normal-salt diet [NS, n = 5]; 2) HS diet [8% NaCl, n = 5]; 3) ANG II infusion in NS rats [ANG II 80 ng/min, n = 5]; 4) ANG II infusion in HS rats [ANG II+HS, n = 5]; and 5) ANG II infusion in HS rats treated with ANG II type 1 receptor blocker (ARB) [ANG II+HS+ARB, n = 5] for 14 days. Rats fed a HS diet alone did not show changes in systolic blood pressure (SBP), proteinuria, cell proliferation, or uAGT excretion although they did exhibit mesangial expansion, collagen deposition, and had increased NADPH oxidase activity accompanied by increased peroxynitrite formation in the kidneys. Compared with ANG II rats, the combination of ANG II infusion and a HS diet led to exacerbation in SBP (175 ± 10 vs. 221 ± 8 mmHg; P < 0.05), proteinuria (46 ± 7 vs. 127 ± 7 mg/day; P < 0.05), and uAGT (1,109 ± 70 vs.. 7,200 ± 614 ng/day; P < 0.05) associated with greater collagen deposition, mesangial expansion, interstitial cell proliferation, and macrophage infiltration. In both ANG II groups, the O(2)(-) levels were increased due to increased NADPH oxidase activity without concomitant increases in peroxynitrite formation. The responses in ANG II rats were prevented or ameliorated by ARB treatment. The results indicate that HS independently stimulates ROS formation, which may synergize with the effect of ANG II to limit peroxynitrite formation, leading to exacerbation of uAGT and greater injury during ANG II salt hypertension.


Subject(s)
Angiotensinogen/biosynthesis , Hypertension/physiopathology , Kidney/drug effects , Kidney/physiopathology , Oxidative Stress/drug effects , Receptor, Angiotensin, Type 1/physiology , Sodium Chloride, Dietary/administration & dosage , Angiotensin II , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensinogen/urine , Animals , Hypertension/chemically induced , Hypertension/pathology , Kidney/pathology , Male , NADPH Oxidases/metabolism , Peroxynitrous Acid/biosynthesis , Proteinuria/etiology , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/pharmacology
10.
Ann Surg Oncol ; 19(6): 1897-901, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22207049

ABSTRACT

BACKGROUND: In adolescents and young adults, primary hyperparathyroidism (PHPT) is an uncommon diagnosis. We compared the clinical characteristics of these patients to those of older adult patients with PHPT. We hypothesized that PHPT in adolescents and young adults is more often caused by single-gland disease and is amenable to minimally invasive parathyroidectomy (MIP). METHODS: We retrospectively reviewed the medical records of 452 consecutive patients who had surgery for PHPT. Patients ranged in age from 13 to 94 years and were dichotomized into younger (age <30 years, n = 17, 3.8%) and older (age ≥ 30 years, n = 435, 96.2%) patients. Continuous baseline and intraoperative and postoperative measures were not normally distributed and were summarized with medians and interquartile ranges (IQRs). Groups were compared using Wilcoxon rank sum test or Fisher exact test, and significance was set at P < .05. RESULTS: Median [IQR] age was 24 [23-27] years for the younger group and 58 [51-66] years for the older group. Though not statistically significant, a smaller proportion of the younger patients compared with the older patients had a positive (99m)Tc-sestamibi scan (71%; 95% confidence interval [95% CI] = 44-90% vs. 83%; 95% CI = 79-86%) and showed a suspected parathyroid adenoma on ultrasound (65%; 95% CI = 38-86% vs. 80%; 95% CI = 76-83%). The younger and older age groups did not significantly differ on preoperative serum PTH levels (median [IQR]: 111 [76-145] pg/ml vs 110 [84-152] pg/ml; P = .73 respectively). The younger group had higher serum calcium levels (11.6 [11.1-12.2] mg/dl) compared with the older group (11.1 [10.7-11.5] mg/dl; P = .01). MIP was performed less frequently on the younger patients (70.6%) compared with older patients (88.7%; P = 0.04). Though the incidence of a single adenoma was somewhat more frequent in older patients (90%; 95% CI = 87-93%) than in younger patients (82%; 95% CI = 57-96%) it was the most frequent cause of PHPT in the younger patients. The younger and older groups did not significantly differ on percent drop from baseline for intraoperative PTH monitoring (81.7 vs 79.3%; P = .46), respectively. CONCLUSIONS: Younger patients with PHPT present with significantly higher serum calcium levels than older patients. However, younger patients are less likely to localize abnormal parathyroid glands on sestamibi or ultrasound. Though younger patients appear to have a higher incidence of hyperplasia compared with older patients, single gland disease is still the overall most frequent cause. Our data suggest that MIP should be more frequently considered in younger patients because of the high incidence of single gland disease.


Subject(s)
Hyperparathyroidism, Primary/surgery , Hyperplasia/surgery , Parathyroid Neoplasms/surgery , Parathyroidectomy , Adolescent , Adult , Age Factors , Aged , Calcium/blood , Female , Follow-Up Studies , Humans , Hyperparathyroidism, Primary/blood , Hyperplasia/blood , Male , Middle Aged , Parathyroid Neoplasms/blood , Postoperative Period , Retrospective Studies , Treatment Outcome , Young Adult
11.
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
12.
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
13.
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
14.
Am J Physiol Renal Physiol ; 299(1): F217-24, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410217

ABSTRACT

Systemic infusion of TNF-alpha exerts renal vasoconstriction but caused marked natriuresis in mice. Similar renal responses were also observed during systemic infusion of nitric oxide (NO) synthase inhibitors as opposed to their usual antinatriuretic responses when administered intrarenally. In the present study, we examined the hypothesis that acute NO blockade systemically induces TNF-alpha generation. which induces this natriuretic response. Renal responses to intravenous infusion of the NO synthase inhibitor nitro-L-arginine methyl ester (L-NAME; 0.2 microg x min(-1) x g body wt(-1) for 85 min) and its impact on the plasma level of TNF-alpha were evaluated in anesthetized mice. Plasma TNF-alpha was undetected in untreated mice (n = 7) but was elevated in L-NAME-treated mice (109 +/- 22 pg/ml; P < 0.01 vs. untreated group; n = 7) along with an increase in TNF-alpha protein expression in kidney tissue. L-NAME infusion caused a usual increase in mean arterial pressure (MAP; 98 +/- 3 to 122 +/- 3 mmHg; P < 0.01) and decreases in renal blood flow (RBF; 8.6 +/- 0.3 to 4.4 +/- 0.2 ml x min(-1) x g(-1); P < 0.01) and glomerular filtration rate (GFR; 1.14 +/- 0.07 to 0.77 +/- 0.04 ml x min(-1) x g(-1); P < 0.01) with a marked increase in sodium excretion (U(Na)V; 0.48 +/- 0.10 to 3.52 +/- 0.85 micromol x min(-1) x g(-1); P < 0.01). Interestingly, in mice (n = 7) pretreated with the TNF-alpha blocker etanercept (5 mg/kg sc), the U(Na)V response to l-NAME infusion was markedly blunted (0.58 +/- 0.08 to 1.22 +/- 0.28 micromol x min(-1) x g(-1); P = NS) although responses for MAP, RBF, and GFR were mostly unchanged. However, pretreatment with the superoxide scavenger tempol in mice (n = 7) did not alter the U(Na)V response to L-NAME. These data demonstrate that L-NAME-induced natriuresis is mediated, at least in part, by concomitant generation of TNF-alpha during NO blockade.


Subject(s)
Anesthesia, General , Enzyme Inhibitors/administration & dosage , Kidney/drug effects , NG-Nitroarginine Methyl Ester/administration & dosage , Natriuresis/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/blood , Animals , Blood Pressure/drug effects , Cyclic N-Oxides/administration & dosage , Etanercept , Free Radical Scavengers/administration & dosage , Glomerular Filtration Rate/drug effects , Immunoglobulin G/administration & dosage , Infusions, Intravenous , Injections, Subcutaneous , Kidney/blood supply , Kidney/enzymology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/metabolism , Receptors, Tumor Necrosis Factor/administration & dosage , Renal Circulation/drug effects , Spin Labels , Superoxides/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation
15.
Am J Physiol Renal Physiol ; 299(3): F656-63, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610532

ABSTRACT

A deficiency in nitric oxide (NO) generation leads to salt-sensitive hypertension, but the role of increased superoxide (O(2)(-)) in such salt sensitivity has not been delineated. We examined the hypothesis that an enhancement in O(2)(-) activity induced by high-salt (HS) intake under deficient NO production contributes to the development of salt-sensitive hypertension. Endothelial NO synthase knockout (eNOS KO; total n = 64) and wild-type (WT; total n = 58) mice were given diets containing either normal (NS; 0.4%) or high-salt (HS; 4%) for 2 wk. During this period, mice were chronically treated with a O(2)(-) scavenger, tempol (400 mg/l), or an inhibitor of NADPH oxidase, apocynin (1 g/l), in drinking water or left untreated (n = 6-8 per group). Blood pressure was measured by radiotelemetry and 24-h urine samples were collected in metabolic cages. Basal mean arterial pressure (MAP) in eNOS KO was higher (125 +/- 4 vs. 106 +/- 3 mmHg) compared with WT. Feeding HS diet did not alter MAP in WT but increased it in eNOS KO to 166 +/- 9 mmHg. Both tempol and apocynin treatment significantly attenuated the MAP response to HS in eNOS KO (134 +/- 3 and 139 +/- 4 mmHg, respectively). Basal urinary 8-isoprostane excretion rates (U(Iso)V), a marker for endogenous O(2)(-) activity, were similar (2.8 +/- 0.2 and 2.4 +/- 0.3 ng/day) in both eNOS KO and WT mice. However, HS increased U(Iso)V more in eNOS KO than in WT (4.6 +/- 0.3 vs. 3.8 +/- 0.2 ng/day); these were significantly attenuated by both tempol and apocynin treatment. These data indicate that an enhancement in O(2)(-) activity contributes substantially to the development of salt-sensitive hypertension under NO-deficient conditions.


Subject(s)
Hypertension/etiology , Hypertension/metabolism , Nitric Oxide Synthase Type III/metabolism , Sodium Chloride, Dietary/pharmacology , Superoxides/metabolism , Acetophenones/pharmacology , Animals , Antioxidants/pharmacology , Blood Pressure/drug effects , Cyclic N-Oxides/pharmacology , Dinoprost/analogs & derivatives , Dinoprost/urine , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Sodium Chloride, Dietary/adverse effects , Spin Labels
16.
Am J Physiol Renal Physiol ; 298(2): F401-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923406

ABSTRACT

To examine the functional interaction between superoxide dismutase (SOD) and NADPH oxidase activity, we assessed renal responses to acute intra-arterial infusion of ANG II (0.5 ng x kg(-1) x min(-1)) before and during administration of a SOD inhibitor, diethyldithiocarbamate (DETC, 0.5 mg x kg(-1) x min(-1)), in enalaprilat-pretreated (33 microg x kg(-1) x min(-1)) rats (n = 11). Total (RBF) and regional (cortical, CBF; medullary; MBF) renal blood flows were determined by Transonic and laser-Doppler flowmetry, respectively. Renal cortical and medullary tissue NADPH oxidase activity in vitro was determined using the lucigenin-chemiluminescence method. DETC treatment alone resulted in decreases in RBF, CBF, MBF, glomerular filtration rate (GFR), urine flow (V), and sodium excretion (U(Na)V) as reported previously. Before DETC, ANG II infusion decreased RBF (-18 +/- 3%), CBF (-16 +/- 3%), MBF [-5 +/- 6%; P = not significant (NS)], GFR (-31 +/- 4%), V (-34 +/- 2%), and U(Na)V (-53 +/- 3%). During DETC infusion, ANG II also caused similar reductions in RBF (-20 +/- 4%), CBF (-19 +/- 3%), MBF (-2 +/- 2; P = NS), and in GFR (-22 +/- 7%), whereas renal excretory responses (V; -12 +/- 2%; U(Na)V; -24 +/- 4%) were significantly attenuated compared with those before DETC. In in vitro experiments, ANG II (100 muM) enhanced NADPH oxidase activity both in cortical [13,194 +/- 1,651 vs. 20,914 +/- 2,769 relative light units (RLU)/mg protein] and in medullary (21,296 +/- 2,244 vs. 30,597 +/- 4,250 RLU/mg protein) tissue. Application of DETC (1 mM) reduced the basal levels and prevented ANG II-induced increases in NADPH oxidase activity in both tissues. These results demonstrate that renal excretory responses to acute ANG II administration are attenuated during SOD inhibition, which seems related to a downregulation of NADPH oxidase in the deficient condition of SOD activity.


Subject(s)
Angiotensin II/administration & dosage , Diuresis/drug effects , Kidney/drug effects , Kidney/metabolism , Superoxide Dismutase/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Dinoprost/analogs & derivatives , Dinoprost/urine , Ditiocarb/administration & dosage , Enalaprilat/administration & dosage , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Infusions, Intra-Arterial , Kidney Cortex/drug effects , Kidney Cortex/enzymology , Male , NADPH Oxidases/metabolism , Nitrates/urine , Nitrites/urine , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects
17.
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.

18.
Am J Physiol Renal Physiol ; 297(6): F1606-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776170

ABSTRACT

Although hypercholesterolemia is implicated in the pathophysiology of many renal disorders as well as hypertension, its direct actions in the kidney are not yet clearly understood. In the present study, we evaluated renal responses to administration of cholesterol (8 microg x min(-1).100 g body wt(-1); bound by polyethylene glycol) into the renal artery of anesthetized male Sprague-Dawley rats. Total renal blood flow (RBF) was measured by a Transonic flow probe, and glomerular filtration rate (GFR) was determined by Inulin clearance. In control rats (n = 8), cholesterol induced reductions of 10 +/- 2% in RBF [baseline (b) 7.6 +/- 0.3 microg x min(-1).100 g(-1)], 17 +/- 3% in urine flow (b, 10.6 +/- 0.9 microg x min(-1).100 g(-1)), 29 +/- 3% in sodium excretion (b, 0.96 +/- 0.05 mumol.min(-1).100 g(-1)) and 24 +/- 2% in nitrite/nitrate excretion (b, 0.22 +/- 0.01 nmol.min(-1).100 g(-1)) without an appreciable change in GFR (b, 0.87 +/- 0.03 ml.min(-1).100 g(-1)). These renal vasoconstrictor and anti-natriuretic responses to cholesterol were absent in rats pretreated with nitric oxide (NO) synthase inhibitor, nitro-l-arginine methylester (0.5 microg x min(-1).100 g(-1); n = 6). In rats pretreated with superoxide (O(2)(-)) scavenger tempol (50 microg x min(-1).100 g(-1); n = 6), the cholesterol-induced renal responses remained mostly unchanged, although there was a slight attenuation in anti-natriuretic response. This anti-natriuretic response to cholesterol was abolished in furosemide-pretreated rats (0.3 microg x min(-1).100 g(-1); n = 6) but remained unchanged in amiloride-pretreated rats (0.2 microg x min(-1).100 g(-1); n = 5), indicating that Na(+)/K(+)/2Cl(-) cotransport is the dominant mediator of this effect. These data demonstrate that cholesterol-induced acute renal vasoconstrictor and antinatriuretic responses are mediated by a decrease in NO production. These data also indicate that tubular effect of cholesterol on sodium reabsorption is mediated by the furosemide sensitive Na(+)/K(+)/2Cl(-) cotransporter.


Subject(s)
Cholesterol/administration & dosage , Kidney/blood supply , Kidney/physiology , Natriuresis/drug effects , Nitric Oxide/antagonists & inhibitors , Vasoconstriction/drug effects , Absorption/drug effects , Animals , Antioxidants/pharmacology , Cyclic N-Oxides/pharmacology , Drug Carriers , Enzyme Inhibitors/pharmacology , Furosemide/pharmacology , Hemodynamics/drug effects , Infusions, Intra-Arterial , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Polyethylene Glycols , Rats , Rats, Sprague-Dawley , Renal Artery , Sodium-Potassium-Chloride Symporters/metabolism , Spin Labels
19.
20.
J Hypertens ; 25(11): 2257-65, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17921820

ABSTRACT

OBJECTIVE: The present study was performed to examine the role of superoxide (O2*) and its interaction with nitric oxide (NO) in the regulation of renal function in prehypertensive heterozygous Ren-2 transgenic rats (TGR). METHODS: Renal responses to the O2* scavenger, tempol (150 microg/min per 100 g), and/or the NO synthase inhibitor, nitro-L-arginine methylester (L-NAME; 5 microg/min per 100 g), infused alone or in combination directly into the renal artery were evaluated in anesthetized heterozygous male TGR and aged-matched Hanover Sprague-Dawley rats (HanSD). RESULTS: There were no differences in arterial pressure (122 +/- 3 versus 115 +/- 2 mmHg), renal plasma flow (RPF; 2.09 +/- 0.1 versus 2.07 +/- 0.1 ml/min per g), glomerular filtration rate (GFR; 0.73 +/- 0.1 versus 0.74 +/- 0.1 ml/min per g) or sodium excretion (0.63 +/- 0.13 versus 0.67 +/- 0.16 micromol/min per g) between TGR and HanSD. Tempol alone caused significant increases in RPF and GFR (10 +/- 4% and 12 +/- 2%, respectively) in TGR but not in HanSD. Tempol also caused greater sodium excretory responses in TGR compared to HanSD (112 +/- 16% versus 43 +/- 7%; P < 0.05). 8-Isoprostane excretion was significantly higher in TGR than in HanSD (10.2 +/- 0.8 versus 6.5 +/- 0.7 pg/min per g), which was attenuated by tempol. L-NAME caused greater decreases in RPF and GFR in TGR (-34 +/- 4% and -22 +/- 4%, respectively) than in HanSD (-19 +/- 3% and -10 +/- 4%, respectively). Co-infusion of tempol partially attenuated the renal hemodynamic and excretory responses to L-NAME in TGR. CONCLUSIONS: These data suggest that the enhanced O2* activity and its interaction with NO during the prehypertensive phase in TGR modulates renal hemodynamic and excretory function, which may contribute to the development of hypertension in this transgenic rat model.


Subject(s)
Hypertension/etiology , Kidney/physiology , Nitric Oxide/physiology , Renin/genetics , Superoxides/metabolism , Animals , Animals, Genetically Modified , Blood Pressure , Cyclic N-Oxides/pharmacology , Glomerular Filtration Rate/drug effects , Hypertension/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Sodium/urine , Spin Labels
SELECTION OF CITATIONS
SEARCH DETAIL