1
|
Cowley AW, Roman RJ, Mattson DL, Franchini KG, O'Connor PM, Makino A, Taylor NE, Evans LC, Mori T, Dickhout JG, Jin C, Miyata N, Nakanishi K, Szentiványi M, Park F, Skelton MM, Kurth T, Shimada S. Renal Medulla in Hypertension. Hypertension 2024; 81:2383-2394. [PMID: 39344517 PMCID: PMC11578791 DOI: 10.1161/hypertensionaha.124.21711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Studies have found that blood flow to the renal medulla is an important determinant of pressure-natriuresis and the long-term regulation of arterial pressure. First, a brief review of methods developed enabling the study of the medullary circulation is presented. Second, studies performed in rats are presented showing medullary blood flow plays a vital role in the pressure-natriuresis relationship and thereby in hypertension. Third, it is shown that chronic reduction of medullary blood flow results in hypertension and that enhancement of medullary blood flow reduces hypertension hereditary models of both salt-sensitive rats and salt-resistant forms of hypertension. The key role that medullary nitric oxide production plays in protecting this region from ischemic injury associated with circulating vasoconstrictor agents and reactive oxygen species is presented. The studies cited are largely the work of my students, research fellows, and colleagues with whom I have performed these studies dating from the late 1980s to more recent years.
Collapse
Affiliation(s)
- Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
| | | | | | | | | | - Ayako Makino
- Herbert Wertheim University of Florida Scripps Institute; University of Florida, Jupsiter (A.M.)
| | | | | | - Takefumi Mori
- Tohoku Medical and Pharmaceutical University, Sendai, Japan (T.M.)
| | | | - Chunhua Jin
- University of Alabama School of Medicine, Birmingham (C.J.)
| | - Noriyuki Miyata
- Taisho Pharmaceutical Co, Ltd. Holdings, Tokyo, Japan (N.M.)
| | | | | | | | - Meredith M Skelton
- Department of Physiology, Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
| | - Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
- Medical College of Wisconsin, Milwaukee (A.W.C., M.M.S., T.K., S.S.)
| |
Collapse
|
2
|
Betrie AH, Ma S, Ow CPC, Peiris RM, Evans RG, Ayton S, Lane DJR, Southon A, Bailey SR, Bellomo R, May CN, Lankadeva YR. Renal arterial infusion of tempol prevents medullary hypoperfusion, hypoxia, and acute kidney injury in ovine Gram-negative sepsis. Acta Physiol (Oxf) 2023; 239:e14025. [PMID: 37548350 PMCID: PMC10909540 DOI: 10.1111/apha.14025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023]
Abstract
AIM Renal medullary hypoperfusion and hypoxia precede acute kidney injury (AKI) in ovine sepsis. Oxidative/nitrosative stress, inflammation, and impaired nitric oxide generation may contribute to such pathophysiology. We tested whether the antioxidant and anti-inflammatory drug, tempol, may modify these responses. METHODS Following unilateral nephrectomy, we inserted renal arterial catheters and laser-Doppler/oxygen-sensing probes in the renal cortex and medulla. Noanesthetized sheep were administered intravenous (IV) Escherichia coli and, at sepsis onset, IV tempol (IVT; 30 mg kg-1 h-1 ), renal arterial tempol (RAT; 3 mg kg-1 h-1 ), or vehicle. RESULTS Septic sheep receiving vehicle developed renal medullary hypoperfusion (76 ± 16% decrease in perfusion), hypoxia (70 ± 13% decrease in oxygenation), and AKI (87 ± 8% decrease in creatinine clearance) with similar changes during IVT. However, RAT preserved medullary perfusion (1072 ± 307 to 1005 ± 271 units), oxygenation (46 ± 8 to 43 ± 6 mmHg), and creatinine clearance (61 ± 10 to 66 ± 20 mL min-1 ). Plasma, renal medullary, and cortical tissue malonaldehyde and medullary 3-nitrotyrosine decreased significantly with sepsis but were unaffected by IVT or RAT. Consistent with decreased oxidative/nitrosative stress markers, cortical and medullary nuclear factor-erythroid-related factor-2 increased significantly and were unaffected by IVT or RAT. However, RAT prevented sepsis-induced overexpression of cortical tissue tumor necrosis factor alpha (TNF-α; 51 ± 16% decrease; p = 0.003) and medullary Thr-495 phosphorylation of endothelial nitric oxide synthase (eNOS; 63 ± 18% decrease; p = 0.015). CONCLUSIONS In ovine Gram-negative sepsis, renal arterial infusion of tempol prevented renal medullary hypoperfusion and hypoxia and AKI and decreased TNF-α expression and uncoupling of eNOS. However, it did not affect markers of oxidative/nitrosative stress, which were significantly decreased by Gram-negative sepsis.
Collapse
Affiliation(s)
- Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Shuai Ma
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Division of Nephrology, Shanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Rachel M. Peiris
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Biomedicine Discovery Institute and Department of PhysiologyMonash UniversityMelbourneVictoriaAustralia
| | - Scott Ayton
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Darius J. R. Lane
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Adam Southon
- Translational Neurodegeneration Laboratory, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - Simon R. Bailey
- Faculty of Veterinary and Agricultural SciencesThe University of MelbourneMelbourneVictoriaAustralia
| | - Rinaldo Bellomo
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
- Australian and New Zealand Intensive Care Research CentreMonash UniversityMelbourneVictoriaAustralia
- Department of Intensive CareAustin HospitalMelbourneVictoriaAustralia
- Department of Intensive CareRoyal Melbourne HospitalMelbourneVictoriaAustralia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
- Department of Critical Care, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Kowsar R, Rahimi AM, Sroka M, Mansouri A, Sadeghi K, Bonakdar E, Kateb SF, Mahdavi AH. Risk of mortality in COVID-19 patients: a meta- and network analysis. Sci Rep 2023; 13:2138. [PMID: 36747045 PMCID: PMC9901837 DOI: 10.1038/s41598-023-29364-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Understanding the most relevant hematological/biochemical characteristics, pre-existing health conditions and complications in survivors and non-survivor will aid in predicting COVID-19 patient mortality, as well as intensive care unit (ICU) referral and death. A literature review was conducted for COVID-19 mortality in PubMed, Scopus, and various preprint servers (bioRxiv, medRxiv and SSRN), with 97 observational studies and preprints, consisting of survivor and non-survivor sub-populations. This meta/network analysis comprised 19,014 COVID-19 patients, consisting of 14,359 survivors and 4655 non-survivors. Meta and network analyses were performed using META-MAR V2.7.0 and PAST software. The study revealed that non-survivors of COVID-19 had elevated levels of gamma-glutamyl transferase and creatinine, as well as a higher number of neutrophils. Non-survivors had fewer lymphocytes and platelets, as well as lower hemoglobin and albumin concentrations. Age, hypertension, and cerebrovascular disease were shown to be the most influential risk factors among non-survivors. The most common complication among non-survivors was heart failure, followed by septic shock and respiratory failure. Platelet counts, creatinine, aspartate aminotransferase, albumin, and blood urea nitrogen levels were all linked to ICU admission. Hemoglobin levels preferred non-ICU patients. Lower levels of hemoglobin, lymphocytes, and albumin were associated with increased mortality in ICU patients. This meta-analysis showed that inexpensive and fast biochemical and hematological tests, as well as pre-existing conditions and complications, can be used to estimate the risk of mortality in COVID-19 patients.
Collapse
Affiliation(s)
- Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Amir Mohammad Rahimi
- Department of Developmental Biology, Göttingen Center for Molecular Biosciences (GZMB), Georg-August-University, 37073, Göttingen, Germany
| | - Magdalena Sroka
- University Medical Center Göttingen, Georg-August-University, 37075, Göttingen, Germany
| | - Alireza Mansouri
- Global Agromedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Khaled Sadeghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Elham Bonakdar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | | | - Amir Hossein Mahdavi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| |
Collapse
|
4
|
Liu R, Juncos LA, Lu Y, Wei J, Zhang J, Wang L, Lai EY, Carlstrom M, Persson AEG. The Role of Macula Densa Nitric Oxide Synthase 1 Beta Splice Variant in Modulating Tubuloglomerular Feedback. Compr Physiol 2023; 13:4215-4229. [PMID: 36715280 PMCID: PMC9990375 DOI: 10.1002/cphy.c210043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Abnormalities in renal electrolyte and water excretion may result in inappropriate salt and water retention, which facilitates the development and maintenance of hypertension, as well as acid-base and electrolyte disorders. A key mechanism by which the kidney regulates renal hemodynamics and electrolyte excretion is via tubuloglomerular feedback (TGF), an intrarenal negative feedback between tubules and arterioles. TGF is initiated by an increase of NaCl delivery at the macula densa cells. The increased NaCl activates luminal Na-K-2Cl cotransporter (NKCC2) of the macula densa cells, which leads to activation of several intracellular processes followed by the production of paracrine signals that ultimately result in a constriction of the afferent arteriole and a tonic inhibition of single nephron glomerular filtration rate. Neuronal nitric oxide (NOS1) is highly expressed in the macula densa. NOS1β is the major splice variant and accounts for most of NO generation by the macula densa, which inhibits TGF response. Macula densa NOS1β-mediated modulation of TGF responses plays an essential role in control of sodium excretion, volume and electrolyte hemostasis, and blood pressure. In this article, we describe the mechanisms that regulate macula densa-derived NO and their effect on TGF response in physiologic and pathologic conditions. © 2023 American Physiological Society. Compr Physiol 13:4215-4229, 2023.
Collapse
Affiliation(s)
- Ruisheng Liu
- Department of Molecular Pharmacology & Physiology
- Hypertension and Kidney Research Center, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Luis A. Juncos
- Department of Internal Medicine, Central Arkansas Veterans Healthcare System, Little Rock, AR
| | - Yan Lu
- Division of Nephrology, University of Alabama at Birmingham, Birmingham AL
| | - Jin Wei
- Department of Molecular Pharmacology & Physiology
| | - Jie Zhang
- Department of Molecular Pharmacology & Physiology
| | - Lei Wang
- Department of Molecular Pharmacology & Physiology
| | - En Yin Lai
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Mattias Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - A. Erik G Persson
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Kuczeriszka M, Wąsowicz K. Animal models of hypertension: The status of nitric oxide and oxidative stress and the role of the renal medulla. Nitric Oxide 2022; 125-126:40-46. [PMID: 35700961 DOI: 10.1016/j.niox.2022.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/22/2022] [Accepted: 06/09/2022] [Indexed: 12/21/2022]
Abstract
Hypertension significantly contributes to overall morbidity and mortality worldwide, and animal models of hypertension provide important tools to verify the physiological and molecular mechanisms underlying the development of the disease. A review of the most important models available would provide an insight into the appropriate targets to be addressed in the treatment of different forms of human hypertension. In the animal models discussed a special attention is given to the status and pathophysiological role of nitric oxide and its interaction with reactive oxygen species and oxidative stress. Another focus of the review are the processes running in the renal medulla which are still insufficiently explored. Deficient nitric oxide synthesis and its reduced bioavailability are important determinants of hypertension since NO is recognized as a major control factor of vascular tone homeostasis. For decades perfusion of the renal medulla has also been regarded as one of the blood pressure control factors and, noteworthily, the renal medulla belongs to the tissues with the highest NO content. The list of most often applied animal hypertension models reviewed here includes variants of salt-induced hypertension, the models with genetic background: such as spontaneously hypertensive rats (SHR) and Dahl salt sensitive (SS/SR) rats, Goldblatt 2K-1C hypertensive rats, and also the pharmacologically-plus-dietary salt-induced model known as DOCA-salt hypertension.
Collapse
Affiliation(s)
- Marta Kuczeriszka
- Department of Renal and Body Fluid Physiology, M. Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, A. Pawinskiego 5, Poland.
| | - Krzysztof Wąsowicz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Oczapowskiego 13, Poland
| |
Collapse
|
6
|
Kuczeriszka M, Dobrowolski L, Walkowska A, Baranowska I, Sitek JD, Kompanowska-Jezierska E. Role of Ang1-7 in renal haemodynamics and excretion in streptozotocin diabetic rats. Clin Exp Pharmacol Physiol 2021; 49:432-441. [PMID: 34870864 DOI: 10.1111/1440-1681.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 11/12/2021] [Accepted: 11/24/2021] [Indexed: 11/28/2022]
Abstract
The contribution of angiotensin (1-7) (Ang1-7) to control of extrarenal and renal function may be modified in diabetes. We investigated the effects of Ang1-7 supplementation on blood pressure, renal circulation and intrarenal reactivity (IVR) to vasoactive agents in normoglycaemic (NG) and streptozotocin diabetic rats (DM). In Sprague Dawley DM and NG rats, 3 weeks after streptozotocin (60 mg/kg i.p.) or solvent injection, Ang1-7 was administered (400 ng/min) over the next 2 weeks using subcutaneously implanted osmotic minipumps. For a period of 5 weeks, blood pressure (BP), 24 h water intake and diuresis were determined weekly. In anaesthetised rats, BP, renal total and cortical (CBF), outer (OMBF) and inner medullary (IMBF) perfusion and urine excretion were determined. To check IVR, a short-time infusion of acetylcholine or norepinephrine was randomly given to the renal artery. Unexpectedly, BP did not differ between NG and DM, and this was not modified by Ang-1-7 supplementation. Baseline IMBF was higher in NG vs. DM, and Ang1-7 treatment did not change it in NG but decreased it in DM. In the latter, Ang1-7 increased cortical IVR to vasoconstrictor and vasodilator stimuli. IMBF decrease after high acetylcholine dose seen in untreated NG was reverted to an increase in Ang1-7 treated rats. Irrespective of the glycaemia level, Ang1-7 did not modify BP. However, it impaired medullary circulation in DM, whereas in NG it rendered the medullary vasculature more sensitive to vasodilators. Possibly, the medullary hypoperfusion in DM was mediated by Ang1-7 activation of angiotensin AT-1 receptors which are upregulated by hyperglycaemia.
Collapse
Affiliation(s)
- Marta Kuczeriszka
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Leszek Dobrowolski
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Walkowska
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Iwona Baranowska
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna D Sitek
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Elżbieta Kompanowska-Jezierska
- Department of Renal and Body Fluid Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
7
|
Dąbkowski K, Kreft E, Sałaga-Zaleska K, Chyła G, Kuchta A, Jankowski M. Redox regulation of hemodynamics response to diadenosine tetraphosphate an agonist of P2 receptors and renal function in diet-induced hypercholesterolemic rats. Physiol Rep 2021; 9:e14888. [PMID: 34110719 PMCID: PMC8191177 DOI: 10.14814/phy2.14888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 11/24/2022] Open
Abstract
Hypercholesterolemia and oxidative stress may lead to disturbances in the renal microvasculature in response to vasoactive agents, including P2 receptors (P2R) agonists. We investigated the renal microvascular response to diadenosine tetraphosphate (Ap4A), an agonist of P2R, in diet‐induced hypercholesteremic rats over 28 days, supplemented in the last 10 days with tempol (2 mM) or DL‐buthionine‐(S,R)‐sulfoximine (BSO, 20 mM) in the drinking water. Using laser Doppler flowmetry, renal blood perfusion in the cortex and medulla (CBP, MBP) was measured during the infusion of Ap4A. This induced a biphasic response in the CBP: a phase of rapid decrease was followed by one of rapid increase extended for 30 min in both the normocholesterolemic and hypercholesterolemic rats. The phase of decreased CBP was not affected by tempol or BSO in either group. Early and extended increases in CBP were prevented by tempol in the hypercholesterolemia rats, while, in the normocholesterolemic rats, only the extended increase in CBP was affected by tempol; BSO prevented extended increase in CBP in normocholesterolemic rats. MBP response is not affected by hypercholesterolemia. The hypercholesterolemic rats were characterized by increased urinary albumin and 8‐isoPGF2α excretion. Moreover, BSO increased the urinary excretion of nephrin in the hypercholesterolemic rats but, similar to tempol, did not affect the excretion of albumin in their urine. The results suggest the important role of redox balance in the extracellular nucleotide regulation of the renal vasculature and glomerular injury in hypercholesterolemia.
Collapse
Affiliation(s)
- Kamil Dąbkowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Ewelina Kreft
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Gabriela Chyła
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
8
|
Zhang C, Fang X, Zhang H, Gao W, Hsu HJ, Roman RJ, Fan F. Genetic susceptibility of hypertension-induced kidney disease. Physiol Rep 2021; 9:e14688. [PMID: 33377622 PMCID: PMC7772938 DOI: 10.14814/phy2.14688] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertension is the second leading cause of end-stage renal disease (ESRD) after diabetes mellitus. The significant differences in the incidence of hypertensive ESRD between different patient populations worldwide and patients with and without family history indicate that genetic determinants play an important role in the onset and progression of this disease. Recent studies have identified genetic variants and pathways that may contribute to the alteration of renal function. Mechanisms involved include affecting renal hemodynamics (the myogenic and tubuloglomerular feedback responses); increasing the production of reactive oxygen species in the tubules; altering immune cell function; changing the number, structure, and function of podocytes that directly cause glomerular damage. Studies with hypertensive animal models using substitution mapping and gene knockout strategies have identified multiple candidate genes associated with the development of hypertension and subsequent renal injury. Genome-wide association studies have implicated genetic variants in UMOD, MYH9, APOL-1, SHROOM3, RAB38, and DAB2 have a higher risk for ESRD in hypertensive patients. These findings provide genetic evidence of potential novel targets for drug development and gene therapy to design individualized treatment of hypertension and related renal injury.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Xing Fang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Huawei Zhang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Wenjun Gao
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Han Jen Hsu
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Richard J. Roman
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Fan Fan
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
9
|
Meade RD, Akerman AP, Notley SR, McGinn R, Poirier P, Gosselin P, Kenny GP. Physiological factors characterizing heat-vulnerable older adults: A narrative review. ENVIRONMENT INTERNATIONAL 2020; 144:105909. [PMID: 32919284 DOI: 10.1016/j.envint.2020.105909] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/24/2020] [Accepted: 06/17/2020] [Indexed: 05/26/2023]
Abstract
More frequent and intense periods of extreme heat (heatwaves) represent the most direct challenge to human health posed by climate change. Older adults are particularly vulnerable, especially those with common age-associated chronic health conditions (e.g., cardiovascular disease, hypertension, obesity, type 2 diabetes, chronic kidney disease). In parallel, the global population is aging and age-associated disease rates are on the rise. Impairments in the physiological responses tasked with maintaining homeostasis during heat exposure have long been thought to contribute to increased risk of health disorders in older adults during heatwaves. As such, a comprehensive overview of the provisional links between age-related physiological dysfunction and elevated risk of heat-related injury in older adults would be of great value to healthcare officials and policy makers concerned with protecting heat-vulnerable sectors of the population from the adverse health impacts of heatwaves. In this narrative review, we therefore summarize our current understanding of the physiological mechanisms by which aging impairs the regulation of body temperature, hemodynamic stability and hydration status. We then examine how these impairments may contribute to acute pathophysiological events common during heatwaves (e.g., heatstroke, major adverse cardiovascular events, acute kidney injury) and discuss how age-associated chronic health conditions may exacerbate those impairments. Finally, we briefly consider the importance of physiological research in the development of climate-health programs aimed at protecting heat-vulnerable individuals.
Collapse
Affiliation(s)
- Robert D Meade
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Ashley P Akerman
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Sean R Notley
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Ryan McGinn
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Paul Poirier
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Pierre Gosselin
- Institut National de Santé Publique du Québec and Université Laval, Québec, Québec, Canada
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada; Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
10
|
Woodman AG, Mah R, Keddie DL, Noble RMN, Holody CD, Panahi S, Gragasin FS, Lemieux H, Bourque SL. Perinatal iron deficiency and a high salt diet cause long-term kidney mitochondrial dysfunction and oxidative stress. Cardiovasc Res 2020; 116:183-192. [PMID: 30715197 DOI: 10.1093/cvr/cvz029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/08/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Perinatal iron deficiency (ID) alters developmental trajectories of offspring, predisposing them to cardiovascular dysfunction in later life. The mechanisms underlying this long-term programming of renal function have not been defined. We hypothesized perinatal ID causes hypertension and alters kidney metabolic function and morphology in a sex-dependent manner in adult offspring. Furthermore, we hypothesized these effects are exacerbated by chronic consumption of a high salt diet. METHODS AND RESULTS Pregnant Sprague Dawley rats were fed either an iron-restricted or replete diet prior to and throughout pregnancy. Adult offspring were fed normal or high salt diets for 6 weeks prior to experimentation at 6 months of age. Blood pressure (BP) was assessed via indwelling catheters in anaesthetized offspring; kidney mitochondrial function was assessed via high-resolution respirometry; reactive oxygen species and nitric oxide were quantified via fluorescence microscopy. Adult males, but not females, exhibited increased systolic BP due to ID (P = 0.01) and high salt intake (P = 0.02). In males, but not in females, medullary mitochondrial content was increased by high salt (P = 0.003), while succinate-dependent respiration was reduced by ID (P < 0.05). The combination of perinatal ID and high salt reduced complex IV activity in the cortex of males (P = 0.01). Perinatal ID increased cytosolic superoxide generation (P < 0.001) concomitant with reduced nitric oxide bioavailability (P < 0.001) in male offspring, while high salt increased mitochondrial superoxide in the medulla (P = 0.04) and cytosolic superoxide within the cortex (P = 0.01). Male offspring exhibited glomerular basement membrane thickening (P < 0.05), increased collagen deposition (P < 0.05), and glomerular hypertrophy (interaction, P = 0.02) due to both perinatal ID and high salt. Female offspring exhibited no alterations in mitochondrial function or morphology due to either high salt or ID. CONCLUSION Perinatal ID causes long-term sex-dependent alterations in renal metabolic function and morphology, potentially contributing to hypertension and increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Andrew G Woodman
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Richard Mah
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Danae L Keddie
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Ronan M N Noble
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Claudia D Holody
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Sareh Panahi
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Ferrante S Gragasin
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Helene Lemieux
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Medicine, University of Alberta, Edmonton, Canada.,Faculty Saint-Jean, University of Alberta, Edmonton, Canada
| | - Stephane L Bourque
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
11
|
Xu N, Jiang S, Persson PB, Persson EAG, Lai EY, Patzak A. Reactive oxygen species in renal vascular function. Acta Physiol (Oxf) 2020; 229:e13477. [PMID: 32311827 DOI: 10.1111/apha.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are produced by the aerobic metabolism. The imbalance between production of ROS and antioxidant defence in any cell compartment is associated with cell damage and may play an important role in the pathogenesis of renal disease. NADPH oxidase (NOX) family is the major ROS source in the vasculature and modulates renal perfusion. Upregulation of Ang II and adenosine activates NOX via AT1R and A1R in renal microvessels, leading to superoxide production. Oxidative stress in the kidney prompts renal vascular remodelling and increases preglomerular resistance. These are key elements in hypertension, acute and chronic kidney injury, as well as diabetic nephropathy. Renal afferent arterioles (Af), the primary resistance vessel in the kidney, fine tune renal hemodynamics and impact on blood pressure. Vice versa, ROS increase hypertension and diabetes, resulting in upregulation of Af vasoconstriction, enhancement of myogenic responses and change of tubuloglomerular feedback (TGF), which further promotes hypertension and diabetic nephropathy. In the following, we highlight oxidative stress in the function and dysfunction of renal hemodynamics. The renal microcirculatory alterations brought about by ROS importantly contribute to the pathophysiology of kidney injury, hypertension and diabetes.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Pontus B. Persson
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | | | - En Yin Lai
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Andreas Patzak
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
12
|
Lee DY, Han K, Yu JH, Park S, Heo JI, Seo JA, Kim NH, Yoo HJ, Kim SG, Kim SM, Choi KM, Baik SH, Park YG, Kim NH. Gamma-glutamyl transferase variability can predict the development of end-stage of renal disease: a nationwide population-based study. Sci Rep 2020; 10:11668. [PMID: 32669624 PMCID: PMC7363906 DOI: 10.1038/s41598-020-68603-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 06/24/2020] [Indexed: 12/17/2022] Open
Abstract
The aim of this study is to investigate whether GGT variability is able to predict the risk of end-stage renal disease (ESRD). The study subjects were Koreans who conducted health exams supported by the Korean National Health Insurance Corporation during 2009-2012 (baseline). After excluding individuals aged < 40 years, heavy alcoholics, or those with histories of chronic liver disease or ESRD, we followed 6,058,995 individuals. We calculated the average successive variability (ASV) of GGT values during the 5 years before the baseline as a parameter of variability. Using Cox proportional analyses, we evaluated the risk of ESRD according to GGT ASV quartiles, defined as the initiation of renal replacement therapy or kidney transplantation, or December 31, 2016. During 38,663,279.3 person-years of follow-up, 12,057 cases of ESRD were identified. Compared with GGT ASV quartile 1, the risk of ESRD was higher in ASV quartiles 3-4 and increased serially, even after adjustment for several metabolic parameters, baseline renal function, presence of comorbidities, low income, and baseline GGT and hemoglobin level. The fully adjusted hazard ratios (95% confidence intervals) of GGT ASV quartiles 3 and 4 were 1.06 (1.01-1.12) and 1.12 (1.06-1.18), respectively. In conclusion, GGT variability is a putative risk factor for ESRD in Koreans.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Biostatics, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sanghyun Park
- Department of Biostatics, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jee-In Heo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seon Mee Kim
- Department of Family Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong Gyu Park
- Department of Biostatics, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, 15355, Republic of Korea.
| |
Collapse
|
13
|
Turbeville HR, Johnson AC, Garrett MR, Dent EL, Sasser JM. Nitric oxide and oxidative stress pathways do not contribute to sex differences in renal injury and function in Dahl SS/Jr rats. Physiol Rep 2020; 8:e14440. [PMID: 32652814 PMCID: PMC7354091 DOI: 10.14814/phy2.14440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
The burden of hypertension in the United States is increasing and yields significant morbidity and mortality, and sex differences in hypertension are widely recognized. Reduced nitric oxide (NO) bioavailability and increased oxidative stress are known to contribute to the pathogenesis of hypertensive renal injury, and but their contributions to sex differences in injury progression of are undefined. Our purpose was to test the hypothesis that male hypertensive rats have accelerated renal injury compared to females and to determine the contributions of the nitric oxide pathway and oxidative stress in these differences. Male and female Dahl SS/Jr rats, a model that spontaneously develops hypertension with age, were allowed to age on a 0.3% NaCl diet until 3 or 6 months of age, at which points blood pressure was measured and plasma, tissue, and urine were collected. While no significant sex differences in blood pressure were present at either time point, renal injury measured by urine protein excretion was more severe (male = 44.9 ± 6; female = 15±3 mg/day/100 g bw, p = .0001), and renal function was reduced (male = 0.48 ± 0.02; female = 0.7 ± 0.03 ml min-1 g-1 kw, p = .001) in males compared to females with age. Both male and female rats exhibited reduced nitric oxide metabolites (3 months: male = 0.65 ± 0.1; female = 0.74 ± 0.3; 6 months: male = 0.16 ± 0.1; female = 0.41 ± 0.1 ml min-1 g-1 kw, p, age = 0.02, p, sex = 0.3). Levels of urinary TBARS were similar (3 months: male = 20±1.5; female = 23±1.8; 6 months: male = 26±4.8; female = 23±4.7µM day g-1 kw, p, age = 0.4, p, sex = 0.9), extracellular superoxide dismutase (EC SOD) mRNA was greater in females (3 months: male = 0.35 ± 0.03; female = 1.4 ± 0.2; 6 months: male = 0.4 ± 0.05; female = 1.3 ± 0.1 normalized counts, p, age = 0.7, p, sex < 0.0001), but EC SOD protein expression was not different (3 months: male = 0.01 ± 0.002; female = 0.01 ± 0.002; 6 months: male = 0.02 ± 0.004; female = 0.01 ± 0.002 relative density, p, age = 0.2, p, sex = 0.8). These data support the presence of significant sex differences in renal injury and function in the Dahl S rat and identify a need for further study into the mechanisms involved.
Collapse
Affiliation(s)
- Hannah R. Turbeville
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Ashley C. Johnson
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Michael R. Garrett
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Elena L. Dent
- Department Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Jennifer M. Sasser
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMSUSA
| |
Collapse
|
14
|
Guan Z, Makled MN, Inscho EW. Purinoceptors, renal microvascular function and hypertension. Physiol Res 2020; 69:353-369. [PMID: 32301620 DOI: 10.33549/physiolres.934463] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Proper renal blood flow (RBF) and glomerular filtration rate (GFR) are critical for maintaining normal blood pressure, kidney function and water and electrolyte homeostasis. The renal microvasculature expresses a multitude of receptors mediating vasodilation and vasoconstriction, which can influence glomerular blood flow and capillary pressure. Despite this, RBF and GFR remain quite stable when arterial pressure fluctuates because of the autoregulatory mechanism. ATP and adenosine participate in autoregulatory control of RBF and GFR via activation of two different purinoceptor families (P1 and P2). Purinoceptors are widely expressed in renal microvasculature and tubules. Emerging data show altered purinoceptor signaling in hypertension-associated kidney injury, diabetic nephropathy, sepsis, ischemia-reperfusion induced acute kidney injury and polycystic kidney disease. In this brief review, we highlight recent studies and new insights on purinoceptors regulating renal microvascular function and renal hemodynamics. We also address the mechanisms underlying renal microvascular injury and impaired renal autoregulation, focusing on purinoceptor signaling and hypertension-induced renal microvascular dysfunction. Interested readers are directed to several excellent and comprehensive reviews that recently covered the topics of renal autoregulation, and nucleotides in kidney function under physiological and pathophysiological conditions (Inscho 2009, Navar et al. 2008, Carlstrom et al. 2015, Vallon et al. 2020).
Collapse
Affiliation(s)
- Z Guan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, South Birmingham, USA.
| | | | | |
Collapse
|
15
|
Yang J, Asico LD, Beitelshees AL, Feranil JB, Wang X, Jones JE, Armando I, Cuevas SG, Schwartz GL, Gums JG, Chapman AB, Turner ST, Boerwinkle E, Cooper-DeHoff RM, Johnson JA, Felder RA, Weinman EJ, Zeng C, Jose PA, Villar VAM. Sorting nexin 1 loss results in increased oxidative stress and hypertension. FASEB J 2020; 34:7941-7957. [PMID: 32293069 DOI: 10.1096/fj.201902448r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/13/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022]
Abstract
Acute renal depletion of sorting nexin 1 (SNX1) in mice results in blunted natriuretic response and hypertension due to impaired dopamine D5 receptor (D5 R) activity. We elucidated the molecular mechanisms for these phenotypes in Snx1-/- mice. These mice had increased renal expressions of angiotensin II type 1 receptor (AT1 R), NADPH oxidase (NOX) subunits, D5 R, and NaCl cotransporter. Basal reactive oxygen species (ROS), NOX activity, and blood pressure (BP) were also higher in Snx1-/- mice, which were normalized by apocynin, a drug that prevents NOX assembly. Renal proximal tubule (RPT) cells from hypertensive (HT) Euro-American males had deficient SNX1 activity, impaired D5 R endocytosis, and increased ROS compared with cells from normotensive (NT) Euro-American males. siRNA-mediated depletion of SNX1 in RPT cells from NT subjects led to a blunting of D5 R agonist-induced increase in cAMP production and decrease in Na+ transport, effects that were normalized by over-expression of SNX1. Among HT African-Americans, three of the 12 single nucleotide polymorphisms interrogated for the SNX1 gene were associated with a decrease in systolic BP in response to hydrochlorothiazide (HCTZ). The results illustrate a new paradigm for the development of hypertension and imply that the trafficking protein SNX1 may be a crucial determinant for hypertension and response to antihypertensive therapy.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Laureano D Asico
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Amber L Beitelshees
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jun B Feranil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaoyan Wang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - John E Jones
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Santiago G Cuevas
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Gary L Schwartz
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - John G Gums
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA.,Department of Community Health and Family Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Arlene B Chapman
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eric Boerwinkle
- Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Robin A Felder
- Department of Pathology, The University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Edward J Weinman
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,The Department of Veterans Affairs, Baltimore, MD, USA
| | - Chunyu Zeng
- Department of Cardiology, Fujian Heart Medical Center, Fujian Medical University Union Hospital, Fujian, P.R.China.,Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Van Anthony M Villar
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
16
|
Saez F, Hong NJ, Cabral PD, Garvin JL. Stretch-Induced Increases in Intracellular Ca Stimulate Thick Ascending Limb O 2- Production and Are Enhanced in Dahl Salt-Sensitive Rats. Hypertension 2019; 75:431-438. [PMID: 31865796 DOI: 10.1161/hypertensionaha.119.13765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mechanical stretch raises intracellular Ca (Cai) in many cell types. Luminal flow-derived stretch stimulates O2- production by thick ascending limbs (THALs). Renal O2- is greater in Dahl salt-sensitive (SS) than salt-resistant (SR) rats. We hypothesized that mechanical stretch stimulates Ca influx via TRPV4 (transient receptor potential vanilloid type 4) which in turn raises Cai in THALs; these increases in Cai are necessary for stretch to augment O2- production; and stretch-stimulated, and therefore flow-induced, O2- production is enhanced in SS compared with SR THALs due to elevated Ca influx and increased Cai. Cai and O2- were measured in SS and SR THALs from rats on normal salt using Fura2-acetoxymethyl ester and dihydroethidium, respectively. Stretch raised Cai in SS by 270.4±48.9 nmol/L and by 123.6±27.0 nmol/L in SR THALs (P<0.02). Removing extracellular Ca eliminated the increases and differences in Cai between strains. Knocking down TRPV4 in SS THALs reduced stretch-induced Cai to SR levels (SS: 92.0±15.9 nmol/L; SR: 123.6±27.0 nmol/L). RN1734, a TRPV4 inhibitor, blunted stretch-elevated Cai by ≈75% and ≈66% in SS (P<0.03) and SR (P<0.04), respectively. Stretch augmented O2- production by 58.6±10.2 arbitrary fluorescent units/min in SS and by 24.4±2.6 arbitrary fluorescent units/min in SR THALs (P<0.05). Removal of extracellular Ca blunted stretch-induced increases in O2- and eliminated differences between strains. RN1734 reduced stretch-induced O2- by ≈70% in SS (P<0.005) and ≈60% in SR (P<0.01). Conclusions are as follows: (1) stretch activates TRPV4, which raises Cai in THALs; (2) the increase in Cai stimulates O2- production; and (3) stretch-induced O2- production is enhanced in SS THALs due to greater increases in Cai.
Collapse
Affiliation(s)
- Fara Saez
- From the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
| | - Nancy J Hong
- From the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
| | - Pablo D Cabral
- From the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
| | - Jeffrey L Garvin
- From the Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
17
|
Jackson EK, Mi Z, Kleyman TR, Cheng D. 8-Aminoguanine Induces Diuresis, Natriuresis, and Glucosuria by Inhibiting Purine Nucleoside Phosphorylase and Reduces Potassium Excretion by Inhibiting Rac1. J Am Heart Assoc 2019; 7:e010085. [PMID: 30608204 PMCID: PMC6404173 DOI: 10.1161/jaha.118.010085] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background 8-Aminoguanosine and 8-aminoguanine are K+-sparing natriuretics that increase glucose excretion. Most effects of 8-aminoguanosine are due to its metabolism to 8-aminoguanine. However, the mechanism by which 8-aminoguanine affects renal function is unknown and is the focus of this investigation. Methods and Results Because 8-aminoguanine has structural similarities with inhibitors of the epithelial sodium channel (ENaC), Na+/H+ exchangers, and adenosine A1 receptors, we examined the effects of 8-aminoguanine on EN aC activity in mouse collecting duct cells, on intracellular pH of human proximal tubular epithelial cells, on responses to a selective A1-receptor agonist in vivo, and on renal excretory function in A1-receptor knockout rats. These experiments showed that 8-aminoguanine did not block EN aC, Na+/H+ exchangers, or A1 receptors. Because Rac1 enhances activity of mineralocorticoid receptors and some guanosine analogues inhibit Rac1, we examined the effects of 8-aminoguanine on Rac1 activity in mouse collecting duct cells. Rac1 activity was significantly inhibited by 8-aminoguanine. Because in vitro 8-aminoguanine is a purine nucleoside phosphorylase ( PNP ase) inhibitor, we examined the effects of a natriuretic dose of 8-aminoguanine on urinary excretion of PNP ase substrates and products. 8-Aminoguanine increased and decreased, respectively, urinary excretion of PNP ase substrates and products. Next we compared in rats the renal effects of intravenous doses of 9-deazaguanine ( PNP ase inhibitor) versus 8-aminoguanine. 8-Aminoguanine and 9-deazaguanine induced similar increases in urinary Na+ and glucose excretion, yet only 8-aminoguanine reduced K+ excretion. Nsc23766 (Rac1 inhibitor) mimicked the effects of 8-aminoguanine on K+ excretion. Conclusions 8-Aminoguanine increases Na+ and glucose excretion by blocking PNP ase and decreases K+ excretion by inhibiting Rac1.
Collapse
Affiliation(s)
- Edwin K Jackson
- 2 Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine Pittsburgh PA
| | - Zaichuan Mi
- 2 Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine Pittsburgh PA
| | - Thomas R Kleyman
- 1 Renal-Electrolyte Division Department of Medicine University of Pittsburgh School of Medicine Pittsburgh PA
| | - Dongmei Cheng
- 2 Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine Pittsburgh PA
| |
Collapse
|
18
|
Yang N, Gonzalez-Vicente A, Garvin JL. Angiotensin II-induced superoxide and decreased glutathione in proximal tubules: effect of dietary fructose. Am J Physiol Renal Physiol 2019; 318:F183-F192. [PMID: 31760771 DOI: 10.1152/ajprenal.00462.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Angiotensin II exacerbates oxidative stress in part by increasing superoxide (O2-) production by many renal tissues. However, whether it does so in proximal tubules and the source of O2- in this segment are unknown. Dietary fructose enhances the stimulatory effect of angiotensin II on proximal tubule Na+ reabsorption, but whether this is true for oxidative stress is unknown. We hypothesized that angiotensin II causes proximal nephron oxidative stress in part by stimulating NADPH oxidase (NOX)4-dependent O2- production and decreasing the amount of the antioxidant glutathione, and this is exacerbated by dietary fructose. We measured basal and angiotensin II-stimulated O2- production with and without inhibitors, NOX1 and NOX4 expression, and total and reduced glutathione (GSH) in proximal tubules from rats drinking either tap water (control) or 20% fructose. Angiotensin II (10 nM) increased O2- production by 113 ± 42 relative light units·mg protein-1·s-1 in controls and 401 ± 74 relative light units·mg protein-1·s-1 with 20% fructose (n = 11 for each group, P < 0.05 vs. control). Apocynin and the Nox1/4 inhibitor GKT136901 prevented angiotensin II-induced increases in both groups. NOX4 expression was not different between groups. NOX1 expression was undetectable. Angiotensin II decreased GSH by 1.8 ± 0.8 nmol/mg protein in controls and by 4.2 ± 0.9 nmol/mg protein with 20% fructose (n = 18 for each group, P < 0.047 vs. control). We conclude that 1) angiotensin II causes oxidative stress in proximal tubules by increasing O2- production by NOX4 and decreasing GSH and 2) dietary fructose enhances the ability of angiotensin II to stimulate O2- and diminish GSH, thereby exacerbating oxidative stress in this segment.
Collapse
Affiliation(s)
- Nianxin Yang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
19
|
de Paula Ribeiro J, Kalb AC, de Bastos Maya S, Gioda A, Martinez PE, Monserrat JM, Jiménez-Vélez BD, Gioda CR. The impact of polar fraction of the fine particulate matter on redox responses in different rat tissues. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:32476-32487. [PMID: 31617135 DOI: 10.1007/s11356-019-06452-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 09/06/2019] [Indexed: 06/10/2023]
Abstract
Particulate matter (PM) contains different chemical substances that have been associated with health effects and an increased risk of mortality due to their toxicity. In this study, fine particulate matter (PM2.5) samples were collected in a region with rural characteristics (Seropédica (Se)) and another with some industries (Duque de Caxias (DC)) (Brazil, RJ). Rats were exposed to PM2.5 extracts daily for 25 days at different dilutions: 10×, 5×, and a concentrated solution (CS). Biochemical analyses were investigated for total antioxidant capacity (ACAP), lipid peroxidation (LPO) levels, reduced glutathione (GSH) concentration, activity of glutamate cysteine ligase (GCL), and activity of glutathione S-transferase (GST). The liver showed a significant increase in GCL (DC-5×, DC-CS and Se-CS) and GST activities (DC-CS and Se-CS) in both regions when compared to the control group. In the renal cortex, GCL activity decreased in most of the tested groups while GST activity increased only in the 5× groups of both regions (DC and Se). In the renal medulla, GCL activity decreased for Se-10× and DC-CS but increased for Se-5×, and GST activity increased in the Se-10×, DC-5×, and DC-CS groups. Lung GCL increased in all groups for both regions. Moreover, this organ also showed an increase in GST activity when higher metal concentrations were present (5× and CS). TBARS levels were increased for all tissues in most tested concentrations. These data indicate that soluble compounds (e.g., metals) from PM2.5 sampled in areas with different pollution indexes can change the redox status and cause damage to different tissues.
Collapse
Affiliation(s)
- Joaquim de Paula Ribeiro
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
- Programa de Pós Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, FURG, Rio Grande, RS, Brazil
| | - Ana Cristina Kalb
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
- Programa de Pós Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, FURG, Rio Grande, RS, Brazil
| | - Sabrina de Bastos Maya
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| | - Adriana Gioda
- Department of Chemistry, Pontifical Catholic University of Rio de Janeiro (PUC-Rio), Rua Marques de São Vicente 225, Gávea, Rio de Janeiro, RJ, 22451-900, Brazil.
| | - Pablo Elias Martinez
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
- Programa de Pós Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, FURG, Rio Grande, RS, Brazil
| | - José Maria Monserrat
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
- Programa de Pós Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, FURG, Rio Grande, RS, Brazil
| | - Braulio D Jiménez-Vélez
- Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Carolina Rosa Gioda
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
- Programa de Pós Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, FURG, Rio Grande, RS, Brazil
| |
Collapse
|
20
|
CCR2 knockout ameliorates obesity-induced kidney injury through inhibiting oxidative stress and ER stress. PLoS One 2019; 14:e0222352. [PMID: 31498850 PMCID: PMC6733486 DOI: 10.1371/journal.pone.0222352] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/27/2019] [Indexed: 12/23/2022] Open
Abstract
CCL2/CCR2 signaling is believed to play an important role in kidney diseases. Several studies have demonstrated that blocking of CCR2 has a therapeutic effect on kidney diseases. However, the effects of CCR2 knockout on obesity-induced kidney injury remain unclear. We investigated the therapeutic effects and the mechanism of CCL2/CCR2 signaling in obesity-induced kidney injury. We used C57BL/6-CCR2 wild type and C57BL/6-CCR2 knockout mice: Regular diet wild type (RD WT), RD CCR2 knockout (RD KO), High-fat diet WT (HFD WT), HFD CCR2 KO (HFD KO). Body weight of WT mice was significantly increased after HFD. However, the body weight of HFD KO mice was not decreased compared to HFD WT mice. Food intake and calorie showed no significant differences between HFD WT and HFD KO mice. Glucose, insulin, total cholesterol, and triglycerides levels increased in HFD WT mice were decreased in HFD KO mice. Insulin resistance, increased insulin secretion, and lipid accumulation showed in HFD WT mice were improved in HFD KO mice. Increased desmin expression, macrophage infiltration, and TNF-α in HFD mice were reduced in HFD KO mice. HFD-induced albuminuria, glomerular hypertrophy, glomerular basement membrane thickening, and podocyte effacement were restored by CCR2 depletion. HFD-induced elevated expressions of xBP1, Bip, and Nox4 at RNA and protein levels were significantly decreased in HFD KO. Therefore, blockade of CCL2/CCR2 signaling by CCR2 depletion might ameliorate obesity-induced albuminuria through blocking oxidative stress, ER stress, and lipid accumulation.
Collapse
|
21
|
Chopra A, Sivaraman K. An update on possible pathogenic mechanisms of periodontal pathogens on renal dysfunction. Crit Rev Microbiol 2019; 45:514-538. [PMID: 30729832 DOI: 10.1080/1040841x.2018.1553847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/17/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022]
Abstract
Periodontitis is a potential source of permanent systemic inflammation that initiates renal dysfunction and contributes to the development of chronic kidney diseases (CKDs). Although numerous studies have confirmed the bidirectional role of periodontal infection and renal inflammation, no literature has yet highlighted the sophisticated pathogenic mechanisms by which periodontal pathogens, particularly Porphynomonas Gingivalis, induce renal dysfunction and contributed in the development of CKDs. The present review aims to critically analyze and highlight the novel pathogenesis of periodontitis induced CKDs.
Collapse
Affiliation(s)
- Aditi Chopra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Karthik Sivaraman
- Department of Prosthodontics, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
22
|
Duni A, Liakopoulos V, Roumeliotis S, Peschos D, Dounousi E. Oxidative Stress in the Pathogenesis and Evolution of Chronic Kidney Disease: Untangling Ariadne's Thread. Int J Mol Sci 2019; 20:ijms20153711. [PMID: 31362427 PMCID: PMC6695865 DOI: 10.3390/ijms20153711] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
Amplification of oxidative stress is present since the early stages of chronic kidney disease (CKD), holding a key position in the pathogenesis of renal failure. Induction of renal pro-oxidant enzymes with excess generation of reactive oxygen species (ROS) and accumulation of dityrosine-containing protein products produced during oxidative stress (advanced oxidation protein products—AOPPs) have been directly linked to podocyte damage, proteinuria, and the development of focal segmental glomerulosclerosis (FSGS) as well as tubulointerstitial fibrosis. Vascular oxidative stress is considered to play a critical role in CKD progression, and ROS are potential mediators of the impaired myogenic responses of afferent renal arterioles in CKD and impaired renal autoregulation. Both oxidative stress and inflammation are CKD hallmarks. Oxidative stress promotes inflammation via formation of proinflammatory oxidized lipids or AOPPs, whereas activation of nuclear factor κB transcription factor in the pro-oxidant milieu promotes the expression of proinflammatory cytokines and recruitment of proinflammatory cells. Accumulating evidence implicates oxidative stress in various clinical models of CKD, including diabetic nephropathy, IgA nephropathy, polycystic kidney disease as well as the cardiorenal syndrome. The scope of this review is to tackle the issue of oxidative stress in CKD in a holistic manner so as to provide a future framework for potential interventions.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Dimitrios Peschos
- Laboratory of Physiology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
23
|
Mohammadi M, Najafi H, Mohamadi Yarijani Z, Vaezi G, Hojati V. Protective effect of piperine in ischemia-reperfusion induced acute kidney injury through inhibition of inflammation and oxidative stress. J Tradit Complement Med 2019; 10:570-576. [PMID: 33134133 PMCID: PMC7588331 DOI: 10.1016/j.jtcme.2019.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/12/2019] [Accepted: 07/25/2019] [Indexed: 11/21/2022] Open
Abstract
Background and aim Renal ischemia-reperfusion is associated with inflammation and oxidative stress. As a major compound in black pepper, piperine has anti-inflammatory and anti-oxidative properties. In present study, the protective effects of oral administration of piperine in renal ischemia-reperfusion (IR) induced acute kidney injuries (AKI) were investigated. Experimental procedure Male Wistar rats received piperine (10 or 20 mg/kg.bw) or vehicle for 10 days. The artery and vein of both kidneys were then clamped for 30 min, followed by a 24-h reperfusion period. Concentrations of creatinine and urea-nitrogen in descending aorta blood were measured, and malondialdehyde (MDA) and ferric reducing/antioxidant power (FRAP) levels were measured in kidney tissue to evaluate the oxidative stress. Inflammation was evaluated by measuring the TNF-α and ICAM-1 mRNA expression levels in renal cortical tissue using Real Time PCR method and counting leukocytes infiltration to interstitium. Further measured were tissue damages in H & E stained sections. Results Renal IR reduced FRAP, while increasing the plasma concentrations of creatinine and urea-nitrogen, tissue MDA level, TNF-α and ICAM-1 mRNA expressions, leukocyte infiltration and histopathologic injuries. Piperine administration significantly reduced the plasma concentrations of creatinine and urea-nitrogen, expression of pro-inflammatory factors, oxidative stress and renal histopathologic injuries. It is to be noted that 20 mg/kg dose was more effective. Conclusion Our results suggest piperine protects the kidney against ischemia-reperfusion induced acute kidney injuries by its anti-inflammatory and anti-oxidative properties. Renal ischemia-reperfusion increased the inflammation and oxidative stress parameters. Ischemia-reperfusion increased histopathological damages and functional parameters. Piperine pretreatment significantly reduced the inflammation and oxidative stress. Piperine administration ameliorated renal function and histopathologic damages.
Collapse
Key Words
- AKI, Acute kidney injury
- Acute kidney injury
- FRAP, Ferric reducing antioxidant power
- GFR, Glomerular filtration rate
- ICAM-1, Intercellular adhesion molecule-1
- IL-1, Interleukin-1
- IL-6, Interleukin-6
- IR, Ischemia-reperfusion
- Inflammation
- Ischemia-reperfusion
- MDA, Malondialdehyde
- NF-κB, Nuclear factor-κB
- NO, Nitric oxide
- Oxidative stress
- PBS, Phosphate buffer saline
- Piperine
- ROS, Reactive oxygen species
- TNF-α, Tumor necrosis factor-α
- TPTZ, Tripyridyl-s-triazine
- eNOS, Endothelial nitric oxide synthase
- iNOS, Inducible nitric oxide synthase
- qRT-PCR, quantitative real-time PCR
Collapse
Affiliation(s)
- Maryam Mohammadi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Houshang Najafi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeynab Mohamadi Yarijani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamhasan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Vida Hojati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
24
|
Balakrishnan K, Panneerpandian P, Devanandan HJ, Sekar BT, Rayala SK, Ganesan K. Salt-mediated transcriptional and proteasomal dysregulations mimic the molecular dysregulations of stomach cancer. Toxicol In Vitro 2019; 61:104588. [PMID: 31279909 DOI: 10.1016/j.tiv.2019.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Karthik Balakrishnan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Ponmathi Panneerpandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Helen Jemimah Devanandan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Balaji T Sekar
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamilnadu, India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamilnadu, India.
| |
Collapse
|
25
|
Haque MZ, Ortiz PA. Superoxide increases surface NKCC2 in the rat thick ascending limbs via PKC. Am J Physiol Renal Physiol 2019; 317:F99-F106. [PMID: 31091128 DOI: 10.1152/ajprenal.00232.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The apical Na+-K+-2Cl- cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb (TAL). The free radical superoxide ( O2- ) stimulates TAL NaCl absorption by enhancing NKCC2 activity. In contrast, nitric oxide (NO) scavenges O2- and inhibits NKCC2. NKCC2 activity depends on the number of NKCC2 transporters in the TAL apical membrane and its phosphorylation. We hypothesized that O2- stimulates NKCC2 activity by enhancing apical surface NKCC2 expression. We measured surface NKCC2 expression in rat TALs by surface biotinylation and Western blot analysis. Treatment of TALs with O2- produced by exogenous xanthine oxidase (1 mU/ml) and hypoxanthine (500 µM) stimulated surface NKCC2 expression by ~18 ± 5% (P < 0.05). O2- -stimulated surface NKCC2 expression was blocked by the O2- scavenger tempol (50 µM). Scavenging H2O2 with 100 U/ml catalase did not block the stimulatory effect of xanthine oxidase-hypoxanthine (22 ± 8% increase from control, P < 0.05). Inhibition of endogenous NO production with Nω-nitro-l-arginine methyl ester enhanced surface NKCC2 expression by 21 ± 6% and, when added together with xanthine oxidase-hypoxanthine, increased surface NKCC2 by 41 ± 10% (P < 0.05). Scavenging O2- with superoxide dismutase (300 U/ml) decreased this stimulatory effect by 60% (39 ± 4% to 15 ± 10%, P < 0.05). Protein kinase C inhibition with Gö-6976 (100 nM) blocked O2- -stimulated surface NKCC2 expression (P < 0.05). O2- did not affect NKCC2 phosphorylation at Thr96/101 or its upstream kinases STE20/SPS1-related proline/alanine-rich kinase-oxidative stress-responsive kinase 1. We conclude that O2- increases surface NKCC2 expression by stimulating protein kinase C and that this effect is blunted by endogenous NO. O2- -stimulated apical trafficking of NKCC2 may be involved in the enhanced surface NKCC2 expression observed in Dahl salt-sensitive rats.
Collapse
Affiliation(s)
- Mohammed Ziaul Haque
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| | - Pablo A Ortiz
- Department of Internal Medicine, Hypertension and Vascular Research, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
26
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
27
|
Abdulmahdi W, Rabadi MM, Jules E, Marghani Y, Marji N, Leung J, Zhang F, Siani A, Siskind T, Vedovino K, Chowdhury N, Sekulic M, Ratliff BB. Kidney dysfunction in the low-birth weight murine adult: implications of oxidative stress. Am J Physiol Renal Physiol 2018; 315:F583-F594. [DOI: 10.1152/ajprenal.00164.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Maternal undernutrition (MUN) during pregnancy leads to low-birth weight (LBW) neonates that have a reduced kidney nephron endowment and higher morbidity as adults. Using a severe combined caloric and protein-restricted mouse model of MUN to generate LBW mice, we examined the progression of renal insufficiency in LBW adults. Through 6 mo of age, LBW males experienced greater albuminuria (ELISA analysis), a more rapid onset of glomerular hypertrophy, and a worse survival rate than LBW females. In contrast, both sexes experienced a comparable progressive decline in renal vascular density (immunofluorescence analysis), renal blood flow (Laser-Doppler flowmetry analysis), glomerular filtration rate (FITC-sinistrin clearance analysis), and a progressive increase in systemic blood pressure (measured via tail-cuff method). Isolated aortas from both LBW sexes demonstrated reduced vasodilation in response to ACh, indicative of reduced nitric oxide bioavailability and endothelial dysfunction. ELISA and immunofluorescence analysis revealed a significant increase of circulating reactive oxygen species and NADPH oxidase type 4 (NOX4) expression in both LBW sexes, although these increases were more pronounced in males. Although more effective in males, chronic tempol treatment did improve all observed pathologies in both sexes of LBW mice. Chronic NOX4 inhibition with GKT137831 was more effective than tempol in preventing pathologies in LBW males. In conclusion, despite some minor differences, LBW female and male adults have a reduced nephron endowment comparable with progressive renal and vascular dysfunction, which is associated with increased oxidative stress and subsequent endothelial dysfunction. Tempol treatment and/or NOX4 inhibition attenuates renal and vascular dysfunction in LBW adults.
Collapse
Affiliation(s)
- Wasan Abdulmahdi
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| | - May M. Rabadi
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Edson Jules
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Yara Marghani
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Noor Marji
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Jessica Leung
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Frank Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Avi Siani
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Tamar Siskind
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Kiara Vedovino
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Nazrul Chowdhury
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Miroslav Sekulic
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brian B. Ratliff
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| |
Collapse
|
28
|
AHMEDA AF, RAE MG, ANWEIGI LM, AL OTAIBI MF, AL-MASRI AA, JOHNS EJ. The Effect of Superoxide Dismutase Enzyme Inhibition on Renal Microcirculation of Spontaneously Hypertensive-Stroke Prone and Wistar Rats. Physiol Res 2018; 67:535-541. [DOI: 10.33549/physiolres.933655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
A significant factor in the development of hypertension may be excessive vasoconstriction within the renal medulla. This study therefore investigated the role of superoxide dismutase (SOD) in the regulation of renal medullary and cortical blood perfusion (MBP and CBP, respectively) in both stroke-prone spontaneously hypertensive rats (SHRSP) and normotensive Wistar rats. CBP and MBP were measured before and after intra-renal infusion of the SOD inhibitor, diethyldithio-carbamic acid (DETC). Under basal conditions, mean arterial pressure was significantly greater in SHRSP than Wistar rats, but both MBP and heart rate (HR) were significantly lower in SHRSP relative to Wistar rats (P<0.05, n=7 in both groups). Infusion of DETC (2 mg/kg/min) into the cortico-medullary border area of the kidney significantly decreased MBP in the SHRSPs (by 28±3 %, n=7, P<0.05), indicating a greater vasoconstriction within this vascular bed. However, DETC also significantly decreased MBP in Wistar rats to a similar extent (24±4 %, n=7, P<0.05). These results suggest that superoxide anions play a significant role in reducing renal vascular compliance within the renal medulla in both normotensive and hypertensive animals, although the responses are not greater in the hypertensive relative to the control animals.
Collapse
Affiliation(s)
- A. F. AHMEDA
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | | | | | | | | | | |
Collapse
|
29
|
Saez F, Hong NJ, Garvin JL. NADPH oxidase 4-derived superoxide mediates flow-stimulated NKCC2 activity in thick ascending limbs. Am J Physiol Renal Physiol 2018; 314:F934-F941. [PMID: 29672130 DOI: 10.1152/ajprenal.00631.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Luminal flow augments Na+ reabsorption in the thick ascending limb more than can be explained by increased ion delivery. This segment reabsorbs 30% of the filtered load of Na+, playing a key role in its homeostasis. Whether flow elevations enhance Na+-K+-2Cl- cotransporter (NKCC2) activity and the second messenger involved are unknown. We hypothesized that raising luminal flow augments NKCC2 activity by enhancing superoxide ([Formula: see text]) production by NADPH oxidase 4 (NOX4). NKCC2 activity was measured in thick ascending limbs perfused at either 5 or 20 nl/min with and without inhibitors of [Formula: see text] production. Raising luminal flow from 5 to 20 nl/min enhanced NKCC2 activity from 4.8 ± 0.9 to 6.3 ± 1.2 arbitrary fluorescent units (AFU)/s. Maintaining flow at 5 nl/min did not alter NKCC2 activity. The superoxide dismutase mimetic manganese (III) tetrakis (4-benzoic acid) porphyrin chloride blunted NKCC2 activity from 3.5 ± 0.4 to 2.5 ± 0.2 AFU/s when flow was 20 nl/min but not 5 nl/min. When flow was 20 nl/min, NKCC2 activity showed no change with time. The selective NOX1/4 inhibitor GKT-137831 blunted NKCC2 activity when thick ascending limbs were perfused at 20 nl/min from 7.2 ± 1.1 to 4.5 ± 0.8 AFU/s but not at 5 nl/min. The inhibitor also prevented luminal flow from elevating [Formula: see text] production. Allopurinol, a xanthine oxidase inhibitor, had no effect on NKCC2 activity when flow was 20 nl/min. Tetanus toxin prevents flow-induced stimulation of NKCC2 activity. We conclude that elevations in luminal flow enhance NaCl reabsorption in thick ascending limbs by stimulating NKCC2 via NOX4 activation and increased [Formula: see text]. NKCC2 activation is primarily the result of insertion of new transporters in the membrane.
Collapse
Affiliation(s)
- Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Nancy J Hong
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW This review aims to highlight recent advances on the role of hyperbilirubinemia in hypertension and chronic kidney disease, with a focus on the pathophysiological mechanisms explaining the protective effects of bilirubin. An overview of pharmacologic induction of hyperbilirubinemia will also be discussed. RECENT FINDINGS The findings depict a protective role of bilirubin in the development of hypertension and cardiovascular diseases. Hyperbilirubinemia is also negatively correlated with the development and progression of chronic kidney disease. Commonly used drugs play a role in pharmacologic induction of hyperbilirubinemia. Bilirubin is therefore an exciting target for new therapeutic interventions for its antioxidant properties can be pivotal in the management of hypertension and in preventing and halting the progression of chronic kidney disease. Longitudinal studies are warranted to evaluate the prospective association between bilirubin levels and incident hypertension and chronic kidney disease in the general population. Interventions to induce hyperbilirubinemia need to be explored as a novel therapeutic approach in fighting disease burden.
Collapse
Affiliation(s)
- Ibrahim Mortada
- American University of Beirut Faculty of Medicine, Beirut, Lebanon.
| |
Collapse
|
31
|
Kunutsor SK, Laukkanen JA. Gamma-glutamyltransferase and risk of chronic kidney disease: A prospective cohort study. Clin Chim Acta 2017; 473:39-44. [PMID: 28811239 DOI: 10.1016/j.cca.2017.08.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/03/2017] [Accepted: 08/12/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Elevated serum gamma-glutamyltransferase (GGT) activity has been linked with an increased risk of chronic kidney disease (CKD) in Asian populations. We aimed to assess the prospective association of serum GGT with risk of CKD in a Caucasian population. MATERIALS AND METHODS We related GGT activity to the incidence of CKD in 2338 men aged 42-61years of the Kuopio Ischemic Heart Disease study with normal kidney function at baseline. Repeat measurements of GGT were used to correct for within-person variability. RESULTS During a median follow-up of 25.6years, 221 men developed new-onset CKD. The age-adjusted regression dilution ratio of loge GGT was 0.70 (95% CI: 0.64-0.75). Gamma-glutamyltransferase was log-linearly associated with risk of CKD in age-adjusted analysis. In Cox regression analysis adjusted for age, the hazard ratio (95% CIs) for CKD per standard deviation increase in loge baseline GGT was 1.25 (1.09-1.43) which was attenuated to 1.01 (0.86-1.19) on further adjustment for several confounders. CONCLUSION Contrary to previous evidence of an independent association between elevated GGT and increased risk of CKD in Asian populations, initial evidence of an association between GGT and CKD in Caucasian men was confounded by body mass index, lifestyle factors, and lipids.
Collapse
Affiliation(s)
- Setor K Kunutsor
- School of Clinical Sciences, University of Bristol, Learning & Research Building (Level 1), Southmead Hospital, Southmead Road, Bristol, UK.
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Central Finland Central Hospital, Jyväskylä, Finland
| |
Collapse
|
32
|
Harms JE, Kuczmarski JM, Kim JS, Thomas GD, Kaufman MP. The role played by oxidative stress in evoking the exercise pressor reflex in health and simulated peripheral artery disease. J Physiol 2017; 595:4365-4378. [PMID: 28369936 DOI: 10.1113/jp273816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/23/2017] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS Ligating the femoral artery of a rat for 72 h, a model for peripheral artery disease, causes an exaggerated exercise pressor reflex in response to muscle contraction. Likewise, the hindlimb muscles of rats with ligated femoral arteries show increased levels of reactive oxygen species. Infusion of tiron, a superoxide scavenger, attenuated the exaggerated pressor reflex and reduced reactive oxygen species production in rats with ligated femoral arteries. Conversely, we found no effect of tiron infusion on the pressor reflex in rats with patent femoral arteries. These results suggest a role of reactive oxygen species with respect to causing the exaggerated pressor response to contraction seen in rats with ligated arteries and peripheral artery disease. ABSTRACT Contraction of muscle evokes the exercise pressor reflex (EPR), which is expressed partly by increases in heart rate and arterial pressure. Patients with peripheral artery disease (PAD) show an exaggerated EPR, sometimes report pain when walking and are at risk for cardiac arrthymias. Previous research suggested that reactive oxygen species (ROS) mediate the exaggerated EPR associated with PAD. To examine the effects of ROS on the EPR, we infused a superoxide scavenger, tiron, into the superficial epigastric artery of decerebrated rats. In some, we simulated PAD by ligating a femoral artery for 72 h before the experiment. The peak EPR in 'ligated' rats during saline infusion averaged 31 ± 4 mmHg, whereas the peak EPR in these rats during tiron infusion averaged 13 ± 2 mmHg (n = 12; P < 0.001); the attenuating effect of tiron on the EPR was partly reversed when saline was reinfused into the superficial epigastric artery (21 ± 2 mmHg; P < 0.01 vs. tiron). The peak EPR in 'ligated' rats was also attenuated (n = 7; P < 0.01) by infusion of gp91ds-tat, a peptide that blocks the activity of NAD(P)H oxidase. Tiron infusion had no effect on the EPR in rats with patent femoral arteries (n = 9). Western blots showed that the triceps surae muscles of 'ligated' rats expressed more Nox2 and p67phox, which are components of NADPH oxidase, compared to triceps surae muscles of 'freely perfused' rats. Tiron added to muscle homogenates reduced ROS production in vitro. The results of the present study provide further evidence indicating that ROS mediates the exaggeration of EPR in rats with simulated PAD.
Collapse
Affiliation(s)
- Jonathan E Harms
- Heart and Vascular Institute, Penn State College of Medicine, Hershey, PA, USA
| | | | - Joyce S Kim
- Heart and Vascular Institute, Penn State College of Medicine, Hershey, PA, USA
| | - Gail D Thomas
- Heart and Vascular Institute, Penn State College of Medicine, Hershey, PA, USA
| | - Marc P Kaufman
- Heart and Vascular Institute, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
33
|
Schiffer TA, Friederich-Persson M. Mitochondrial Reactive Oxygen Species and Kidney Hypoxia in the Development of Diabetic Nephropathy. Front Physiol 2017; 8:211. [PMID: 28443030 PMCID: PMC5386984 DOI: 10.3389/fphys.2017.00211] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/23/2017] [Indexed: 12/21/2022] Open
Abstract
The underlying mechanisms in the development of diabetic nephropathy are currently unclear and likely consist of a series of dynamic events from the early to late stages of the disease. Diabetic nephropathy is currently without curative treatments and it is acknowledged that even the earliest clinical manifestation of nephropathy is preceded by an established morphological renal injury that is in turn preceded by functional and metabolic alterations. An early manifestation of the diabetic kidney is the development of kidney hypoxia that has been acknowledged as a common pathway to nephropathy. There have been reports of altered mitochondrial function in the diabetic kidney such as altered mitophagy, mitochondrial dynamics, uncoupling, and cellular signaling through hypoxia inducible factors and AMP-kinase. These factors are also likely to be intertwined in a complex manner. In this review, we discuss how these pathways are connected to mitochondrial production of reactive oxygen species (ROS) and how they may relate to the development of kidney hypoxia in diabetic nephropathy. From available literature, it is evident that early correction and/or prevention of mitochondrial dysfunction may be pivotal in the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Tomas A Schiffer
- Department of Medical Cell Biology, Uppsala UniversityUppsala, Sweden.,Department of Medical and Health Sciences, Linköping UniversityLinköping, Sweden
| | | |
Collapse
|
34
|
Gonzalez-Vicente A, Garvin JL. Effects of Reactive Oxygen Species on Tubular Transport along the Nephron. Antioxidants (Basel) 2017; 6:antiox6020023. [PMID: 28333068 PMCID: PMC5488003 DOI: 10.3390/antiox6020023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 12/17/2022] Open
Abstract
Reactive oxygen species (ROS) are oxygen-containing molecules naturally occurring in both inorganic and biological chemical systems. Due to their high reactivity and potentially damaging effects to biomolecules, cells express a battery of enzymes to rapidly metabolize them to innocuous intermediaries. Initially, ROS were considered by biologists as dangerous byproducts of respiration capable of causing oxidative stress, a condition in which overproduction of ROS leads to a reduction in protective molecules and enzymes and consequent damage to lipids, proteins, and DNA. In fact, ROS are used by immune systems to kill virus and bacteria, causing inflammation and local tissue damage. Today, we know that the functions of ROS are not so limited, and that they also act as signaling molecules mediating processes as diverse as gene expression, mechanosensation, and epithelial transport. In the kidney, ROS such as nitric oxide (NO), superoxide (O₂-), and their derivative molecules hydrogen peroxide (H₂O₂) and peroxynitrite (ONO₂-) regulate solute and water reabsorption, which is vital to maintain electrolyte homeostasis and extracellular fluid volume. This article reviews the effects of NO, O₂-, ONO₂-, and H₂O₂ on water and electrolyte reabsorption in proximal tubules, thick ascending limbs, and collecting ducts, and the effects of NO and O₂- in the macula densa on tubuloglomerular feedback.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1113AAD, Argentina.
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
35
|
Effect of tempol and tempol plus catalase on intra-renal haemodynamics in spontaneously hypertensive stroke-prone (SHSP) and Wistar rats. J Physiol Biochem 2016; 73:207-214. [PMID: 27933463 DOI: 10.1007/s13105-016-0541-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/24/2016] [Indexed: 02/07/2023]
Abstract
Vasoconstriction within the renal medulla contributes to the development of hypertension. This study investigated the role of reactive oxygen species (ROS) in regulating renal medullary and cortical blood perfusion (MBP and CBP respectively) in both stroke-prone spontaneously hypertensive rats (SHRSP) and Wistar rats. CBP and MBP were measured using a laser-Doppler flow meter before and after intra-renal infusion of tempol, the superoxide dismutase (SOD) mimetic or tempol plus catalase, the hydrogen peroxide-degrading enzyme. Tempol infusion significantly elevated blood perfusion within the renal medulla (MBP) in both SHRSP (by 43 ± 7%, P < 0.001) and Wistar rats (by 17 ± 2%, P < 0.05) but the magnitude of the increase was significantly greater in the SHRSP (P < 0.01). When the enzyme catalase and tempol were co-infused, MBP was again significantly increased in SHRSP (by 57 ± 6%, P < 0.001) and Wistar rats (by 33 ± 6%, P < 0.001), with a significantly greater increase in perfusion being induced in the SHRSP relative to the Wistar rats (P < 0.01). Notably, this increase was significantly greater than in those animals infused with tempol alone (P < 0.01). These results suggest that ROS plays a proportionally greater role in reducing renal vascular compliance, particularly within the renal medulla, in normotensive and hypertensive animals, with effects being greater in the hypertensive animals. This supports the hypothesis that SHRSP renal vasculature might be subjected to elevated level of oxidative stress relative to normotensive animals.
Collapse
|
36
|
Persson P, Fasching A, Teerlink T, Hansell P, Palm F. Cellular transport of l-arginine determines renal medullary blood flow in control rats, but not in diabetic rats despite enhanced cellular uptake capacity. Am J Physiol Renal Physiol 2016; 312:F278-F283. [PMID: 27927650 DOI: 10.1152/ajprenal.00335.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 11/22/2022] Open
Abstract
Diabetes mellitus is associated with decreased nitric oxide bioavailability thereby affecting renal blood flow regulation. Previous reports have demonstrated that cellular uptake of l-arginine is rate limiting for nitric oxide production and that plasma l-arginine concentration is decreased in diabetes. We therefore investigated whether regional renal blood flow regulation is affected by cellular l-arginine uptake in streptozotocin-induced diabetic rats. Rats were anesthetized with thiobutabarbital, and the left kidney was exposed. Total, cortical, and medullary renal blood flow was investigated before and after renal artery infusion of increasing doses of either l-homoarginine to inhibit cellular uptake of l-arginine or Nω-nitro- l-arginine methyl ester (l-NAME) to inhibit nitric oxide synthase. l-Homoarginine infusion did not affect total or cortical blood flow in any of the groups, but caused a dose-dependent reduction in medullary blood flow. l-NAME decreased total, cortical and medullary blood flow in both groups. However, the reductions in medullary blood flow in response to both l-homoarginine and l-NAME were more pronounced in the control groups compared with the diabetic groups. Isolated cortical tubular cells displayed similar l-arginine uptake capacity whereas medullary tubular cells isolated from diabetic rats had increased l-arginine uptake capacity. Diabetics had reduced l-arginine concentrations in plasma and medullary tissue but increased l-arginine concentration in cortical tissue. In conclusion, the reduced l-arginine availability in plasma and medullary tissue in diabetes results in reduced nitric oxide-mediated regulation of renal medullary hemodynamics. Cortical blood flow regulation displays less dependency on extracellular l-arginine and the upregulated cortical tissue l-arginine may protect cortical hemodynamics in diabetes.
Collapse
Affiliation(s)
- Patrik Persson
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Angelica Fasching
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Tom Teerlink
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter Hansell
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| | - Fredrik Palm
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden; and
| |
Collapse
|
37
|
Kunutsor SK. Gamma-glutamyltransferase-friend or foe within? Liver Int 2016; 36:1723-1734. [PMID: 27512925 DOI: 10.1111/liv.13221] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/05/2016] [Indexed: 02/13/2023]
Abstract
Gamma-glutamyltransferase (GGT) is a liver enzyme, which is located on the plasma membranes of most cells and organ tissues, but more commonly in hepatocytes, and is routinely used in clinical practice to help indicate liver injury and as a marker of excessive alcohol consumption. Among the liver enzymes, important advances have especially been made in understanding the physiological functions of GGT. The primary role of GGT is the extracellular catabolism of glutathione, the major thiol antioxidant in mammalian cells, which plays a relevant role in protecting cells against oxidants produced during normal metabolism; GGT, therefore, plays an important role in cellular defence. Beyond its physiological functions, circulating serum GGT has been linked to a remarkable array of chronic conditions and diseases, which include nonalcoholic fatty liver disease, vascular and nonvascular diseases and mortality outcomes. This review summarizes the available epidemiological and genetic evidence for the associations between GGT and these adverse outcomes, the postulated biologic mechanisms underlying these associations, outlines areas of outstanding uncertainty and the implications for clinical practice.
Collapse
|
38
|
Dutta UK, Lane J, Roberts LJ, Majid DSA. Superoxide Formation and Interaction with Nitric Oxide Modulate Systemic Arterial Pressure and Renal Function in Salt-Depleted Dogs. Exp Biol Med (Maywood) 2016; 231:269-76. [PMID: 16514172 DOI: 10.1177/153537020623100305] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To determine the role of superoxide (O2–) formation in the kidney during alterations in the renin-angiotensin system, we evaluated responses to the intra-arterial infusion of an O2–-scavenging agent, tempol, in the denervated kidney of anesthetized salt-depleted (SD, n = 6) dogs and salt-replete (SR, n = 6) dogs. As expected, basal plasma renin activity was higher in SD than in SR dogs (8.4 ± 1.0 vs. 2.3 ± 0.6 ng angiotensin 1/ml/hr). Interestingly, the basal level of urinary F2-isoprostanes excretion (marker for endogenous O2– activity) relative to creatinine (Cr) excretion was also significantly higher in SD compared to SR dogs (9.1 ± 2.8 vs. 1.6 ± 0.4 ng F2-isoprostanes/mg of Cr). There was a significant increase in renal blood flow (4.3 ± 0.5 to 4.9 ± 0.6 ml/min/g) and decreases in renal vascular resistance (38.2 ± 5.8 to 33.2 ± 4.7 mm Hg/ml/min/g) and mean systemic arterial pressure (148 ± 6 to 112 ± 10 mm Hg) in SD dogs but not in SR dogs during infusion of tempol at 1 mg/kg/min for 30 mins. Glomerular filtration rate and urinary sodium excretion (UNaV) did not change significantly during tempol infusion in both groups of dogs. Administration of the nitric oxide synthase inhibitor nitro-L-arginine (50 μg/kg/min) during tempol infusion caused a reduction in UNaV in SR dogs (47% ± 12%) but did not cause a decrease in SD dogs. These data show that low salt intake enhances O2– activity that influences renal and systemic hemodynamics and thus may contribute to the regulation of arterial pressure in the salt-restricted state.
Collapse
Affiliation(s)
- Utpal K Dutta
- Department of Physiology, SL 39, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|
39
|
Ngo JP, Ow CP, Gardiner BS, Kar S, Pearson JT, Smith DW, Evans RG. Diffusive shunting of gases and other molecules in the renal vasculature: physiological and evolutionary significance. Am J Physiol Regul Integr Comp Physiol 2016; 311:R797-R810. [DOI: 10.1152/ajpregu.00246.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/27/2016] [Indexed: 01/22/2023]
Abstract
Countercurrent systems have evolved in a variety of biological systems that allow transfer of heat, gases, and solutes. For example, in the renal medulla, the countercurrent arrangement of vascular and tubular elements facilitates the trapping of urea and other solutes in the inner medulla, which in turn enables the formation of concentrated urine. Arteries and veins in the cortex are also arranged in a countercurrent fashion, as are descending and ascending vasa recta in the medulla. For countercurrent diffusion to occur, barriers to diffusion must be small. This appears to be characteristic of larger vessels in the renal cortex. There must also be gradients in the concentration of molecules between afferent and efferent vessels, with the transport of molecules possible in either direction. Such gradients exist for oxygen in both the cortex and medulla, but there is little evidence that large gradients exist for other molecules such as carbon dioxide, nitric oxide, superoxide, hydrogen sulfide, and ammonia. There is some experimental evidence for arterial-to-venous (AV) oxygen shunting. Mathematical models also provide evidence for oxygen shunting in both the cortex and medulla. However, the quantitative significance of AV oxygen shunting remains a matter of controversy. Thus, whereas the countercurrent arrangement of vasa recta in the medulla appears to have evolved as a consequence of the evolution of Henle’s loop, the evolutionary significance of the intimate countercurrent arrangement of blood vessels in the renal cortex remains an enigma.
Collapse
Affiliation(s)
- Jennifer P. Ngo
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| | - Connie P.C. Ow
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| | - Bruce S. Gardiner
- School of Engineering and Information Technology, Murdoch University, Perth, Western Australia
| | - Saptarshi Kar
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - James T. Pearson
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
- Monash Biomedical Imaging Facility, Monash University, Melbourne, Australia
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - David W. Smith
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - Roger G. Evans
- Cardiovascular Disease Program, Biosciences Discovery Institute and Department of Physiology and
| |
Collapse
|
40
|
Gonzalez-Vicente A, Saikumar JH, Massey KJ, Hong NJ, Dominici FP, Carretero OA, Garvin JL. Angiotensin II stimulates superoxide production by nitric oxide synthase in thick ascending limbs. Physiol Rep 2016; 4:4/4/e12697. [PMID: 26884476 PMCID: PMC4759044 DOI: 10.14814/phy2.12697] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Angiotensin II (Ang II) causes nitric oxide synthase (NOS) to become a source of superoxide (O2 (-)) via a protein kinase C (PKC)-dependent process in endothelial cells. Ang II stimulates both NO and O2 (-) production in thick ascending limbs. We hypothesized that Ang II causes O2 (-) production by NOS in thick ascending limbs via a PKC-dependent mechanism. NO production was measured in isolated rat thick ascending limbs using DAF-FM, whereas O2 (-) was measured in thick ascending limb suspensions using the lucigenin assay. Consistent stimulation of NO was observed with 1 nmol/L Ang II (P < 0.001; n = 9). This concentration of Ang II-stimulated O2 (-) production by 50% (1.77 ± 0.26 vs. 2.62 ± 0.36 relative lights units (RLU)/s/μg protein; P < 0.04; n = 5). In the presence of the NOS inhibitor L-NAME, Ang II-stimulated O2 (-) decreased from 2.02 ± 0.29 to 1.10 ± 0.11 RLU/s/μg protein (P < 0.01; n = 8). L-arginine alone did not change Ang II-stimulated O2 (-) (2.34 ± 0.22 vs. 2.29 ± 0.29 RLU/s/μg protein; n = 5). In the presence of Ang II plus the PKC α/β1 inhibitor Gö 6976, L-NAME had no effect on O2 (-) production (0.78 ± 0.23 vs. 0.62 ± 0.11 RLU/s/μg protein; n = 7). In the presence of Ang II plus apocynin, a NADPH oxidase inhibitor, L-NAME did not change O2 (-) (0.59 ± 0.04 vs. 0.61 ± ×0.08 RLU/s/μg protein; n = 5). We conclude that: (1) Ang II causes NOS to produce O2 (-) in thick ascending limbs via a PKC- and NADPH oxidase-dependent process; and (2) the effect of Ang II is not due to limited substrate.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Jagannath H Saikumar
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI
| | - Katherine J Massey
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI
| | - Nancy J Hong
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Fernando P Dominici
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina Instituto de Química y Fisicoquímica Biológicas, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Oscar A Carretero
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio Department of Internal Medicine, Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI
| |
Collapse
|
41
|
La Favor JD, Burnett AL. A microdialysis method to measure in vivo hydrogen peroxide and superoxide in various rodent tissues. Methods 2016; 109:131-140. [PMID: 27452801 DOI: 10.1016/j.ymeth.2016.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 01/07/2023] Open
Abstract
Reactive oxygen species (ROS) play a critical role in cell signaling and disease pathogenesis. Despite their biological importance, assessment of ROS often involves measurement of indirect byproducts or measurement of ROS from excised tissue. Herein, we describe a microdialysis technique that utilizes the Amplex Ultrared assay to directly measure hydrogen peroxide (H2O2) and superoxide in tissue of living, anesthetized rats and mice. We demonstrate the application of this methodology in the penis, adipose tissue, skeletal muscle, kidney, and liver. We provide data demonstrating the impact of important methodological considerations such as membrane length, perfusion rate, and time-dependence upon probe insertion. In this report, we provide a complete list of equipment, troubleshooting tips, and suggestions for implementing this technique in a new system. The data herein demonstrate the feasibility of measuring both in vivo H2O2 and superoxide in the extracellular environment of various rodent tissues, providing a technique with potential application to a vast array of disease states which are subject to oxidative stress.
Collapse
Affiliation(s)
- Justin D La Favor
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltiore, MD, United States.
| | - Arthur L Burnett
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltiore, MD, United States
| |
Collapse
|
42
|
Abstract
SIGNIFICANCE A common link between all forms of acute and chronic kidney injuries, regardless of species, is enhanced generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) during injury/disease progression. While low levels of ROS and RNS are required for prosurvival signaling, cell proliferation and growth, and vasoreactivity regulation, an imbalance of ROS and RNS generation and elimination leads to inflammation, cell death, tissue damage, and disease/injury progression. RECENT ADVANCES Many aspects of renal oxidative stress still require investigation, including clarification of the mechanisms which prompt ROS/RNS generation and subsequent renal damage. However, we currently have a basic understanding of the major features of oxidative stress pathology and its link to kidney injury/disease, which this review summarizes. CRITICAL ISSUES The review summarizes the critical sources of oxidative stress in the kidney during injury/disease, including generation of ROS and RNS from mitochondria, NADPH oxidase, and inducible nitric oxide synthase. The review next summarizes the renal antioxidant systems that protect against oxidative stress, including superoxide dismutase and catalase, the glutathione and thioredoxin systems, and others. Next, we describe how oxidative stress affects kidney function and promotes damage in every nephron segment, including the renal vessels, glomeruli, and tubules. FUTURE DIRECTIONS Despite the limited success associated with the application of antioxidants for treatment of kidney injury/disease thus far, preventing the generation and accumulation of ROS and RNS provides an ideal target for potential therapeutic treatments. The review discusses the shortcomings of antioxidant treatments previously used and the potential promise of new ones. Antioxid. Redox Signal. 25, 119-146.
Collapse
Affiliation(s)
- Brian B Ratliff
- 1 Department of Medicine, Renal Research Institute , New York Medical College, Valhalla, New York.,2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Wasan Abdulmahdi
- 2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Rahul Pawar
- 1 Department of Medicine, Renal Research Institute , New York Medical College, Valhalla, New York
| | - Michael S Wolin
- 2 Department of Physiology, Renal Research Institute , New York Medical College, Valhalla, New York
| |
Collapse
|
43
|
Patschan D, Kribben A, Müller GA. Postischemic microvasculopathy and endothelial progenitor cell-based therapy in ischemic AKI: update and perspectives. Am J Physiol Renal Physiol 2016; 311:F382-94. [PMID: 27194716 DOI: 10.1152/ajprenal.00232.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/15/2016] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) dramatically increases mortality of hospitalized patients. Incidences have been increased in recent years. The most frequent cause is transient renal hypoperfusion or ischemia which induces significant tubular cell dysfunction/damage. In addition, two further events take place: interstitial inflammation and microvasculopathy (MV). The latter evolves within minutes to hours postischemia and may result in permanent deterioration of the peritubular capillary network, ultimately increasing the risk for chronic kidney disease (CKD) in the long term. In recent years, our understanding of the molecular/cellular processes responsible for acute and sustained microvasculopathy has increasingly been expanded. The methodical approaches for visualizing impaired peritubular blood flow and increased vascular permeability have been optimized, even allowing the depiction of tissue abnormalities in a three-dimensional manner. In addition, endothelial dysfunction, a hallmark of MV, has increasingly been recognized as an inductor of both vascular malfunction and interstitial inflammation. In this regard, so-called regulated necrosis of the endothelium could potentially play a role in postischemic inflammation. Endothelial progenitor cells (EPCs), represented by at least two major subpopulations, have been shown to promote vascular repair in experimental AKI, not only in the short but also in the long term. The discussion about the true biology of the cells continues. It has been proposed that early EPCs are most likely myelomonocytic in nature, and thus they may simply be termed proangiogenic cells (PACs). Nevertheless, they reliably protect certain types of tissues/organs from ischemia-induced damage, mostly by modulating the perivascular microenvironment in an indirect manner. The aim of the present review is to summarize the current knowledge on postischemic MV and EPC-mediated renal repair.
Collapse
Affiliation(s)
- D Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Georg-August-University, Göttingen, Germany; and
| | - A Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - G A Müller
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Georg-August-University, Göttingen, Germany; and
| |
Collapse
|
44
|
Saez F, Hong NJ, Garvin JL. Luminal flow induces NADPH oxidase 4 translocation to the nuclei of thick ascending limbs. Physiol Rep 2016; 4:4/6/e12724. [PMID: 27033446 PMCID: PMC4814881 DOI: 10.14814/phy2.12724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/11/2023] Open
Abstract
Superoxide (O2−) exerts its physiological actions in part by causing changes in gene transcription. In thick ascending limbs flow‐induced O2− production is mediated by NADPH oxidase 4 (Nox4) and is dependent on protein kinase C (PKC). Polymerase delta interacting protein 2 (Poldip2) increases Nox4 activity, but it is not known whether Nox4 translocates to the nucleus and whether Poldip2 participates in this process. We hypothesized that luminal flow causes Nox4 translocation to the nuclei of thick ascending limbs in a PKC‐dependent process facilitated by Poldip2. To test our hypothesis, we studied the subcellular localization of Nox4 and Poldip2 using confocal microscopy and O2− production in the absence and presence of luminal flow. Luminal flow increased the ratio of nuclear to cytoplasmic intensity of Nox4 (N/C) from 0.3 ± 0.1 to 0.7 ± 0.1 (P < 0.01) and O2− production from 89 ± 15 to 231 ± 16 AU/s (P < 0.001). In the presence of flow PKC inhibition reduced N/C from 0.5 ± 0.1 to 0.2 ± 0.1 (P < 0.01). Flow‐induced O2− production was also blocked (flow: 142 ± 20 AU/s; flow plus PKC inhibition 26 ± 12 AU/s; P < 0.01). The cytoskeleton disruptor cytochalasin D (1 μmol/L) decreased flow‐induced Nox4 translocation by 0.3 ± 0.01 (P < 0.01); however, it did not reduce flow‐induced O2−. Flow did not alter Poldip2 localization. We conclude that: (1) luminal flow elicits Nox4 translocation to the nucleus in a PKC‐ and cytoskeleton‐dependent process; (2) Nox4 activation occurs before translocation; and (3) Poldip2 is not involved in Nox4 nuclear translocation. Flow‐induced Nox4 translocation to the nucleus may play a role in O2−‐dependent changes in thick ascending limbs.
Collapse
Affiliation(s)
- Fara Saez
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Nancy J Hong
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
45
|
Riyadh Thiab N, King N, McMillan M, Almashhadany A, L Jones G. Age-related protein and mRNA expression of glutathione peroxidases (GPx) and Hsp-70 in different regions of rat kidney with and without stressor. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.2.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
46
|
Cowley AW, Yang C, Zheleznova NN, Staruschenko A, Kurth T, Rein L, Kumar V, Sadovnikov K, Dayton A, Hoffman M, Ryan RP, Skelton MM, Salehpour F, Ranji M, Geurts A. Evidence of the Importance of Nox4 in Production of Hypertension in Dahl Salt-Sensitive Rats. Hypertension 2015; 67:440-50. [PMID: 26644237 DOI: 10.1161/hypertensionaha.115.06280] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/11/2015] [Indexed: 12/24/2022]
Abstract
This study reports the consequences of knocking out NADPH (nicotinamide adenine dinucleotide phosphate) oxidase 4 (Nox4) on the development of hypertension and kidney injury in the Dahl salt-sensitive (SS) rat. Zinc finger nuclease injection of single-cell SS embryos was used to create an 8 base-pair frame-shift deletion of Nox4, resulting in a loss of the ≈68 kDa band in Western blot analysis of renal cortical tissue of the knock out of Nox4 in the SS rat (SS(Nox4-/-)) rats. SS(Nox4-/-) rats exhibited a significant reduction of salt-induced hypertension compared with SS rats after 21 days of 4.0% NaCl diet (134±5 versus 151±3 mm Hg in SS) and a significant reduction of albuminuria, tubular casts, and glomerular injury. Optical fluorescence 3-dimensional cryoimaging revealed significantly higher redox ratios (NADH/FAD [reduced nicotinamide adenine dinucleotide/flavin adenine dinucleotide]) in the kidneys of SS(Nox4-/-) rats even when fed the 0.4% NaCl diet, indicating greater levels of mitochondrial electron transport chain metabolic activity and reduced oxidative stress compared with SS rats. Before the development of hypertension, RNA expression levels of Nox subunits Nox2, p67(phox), and p22(phox) were found to be significantly lower (P<0.05) in SS(Nox4-/-) compared with SS rats in the renal cortex. Thus, the mutation of Nox4 seems to modify transcription of several genes in ways that contribute to the protective effects observed in the SS(Nox4-/-) rats. We conclude that the reduced renal injury and attenuated blood pressure response to high salt in the SS(Nox4-/-) rat could be the result of multiple pathways, including gene transcription, mitochondrial energetics, oxidative stress, and protein matrix production impacted by the knock out of Nox4.
Collapse
Affiliation(s)
- Allen W Cowley
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.).
| | - Chun Yang
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Nadezhda N Zheleznova
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Alexander Staruschenko
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Theresa Kurth
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Lisa Rein
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Vikash Kumar
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Katherine Sadovnikov
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Alex Dayton
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Matthew Hoffman
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Robert P Ryan
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Meredith M Skelton
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Fahimeh Salehpour
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Mahsa Ranji
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| | - Aron Geurts
- From the Department of Physiology (A.W.C., C.Y., N.N.Z., A.S., T.K., V.K., K.S., A.D., M.H., R.P.R., M.M.S., A.G.) and Division of Biostatistics, Institute for Health & Society (L.R.), Medical College of Wisconsin, Milwaukee; and Biophotonics Lab, University of Wisconsin, Milwaukee (F.S., M.R.)
| |
Collapse
|
47
|
Thiab NR, King N, Jones GL. Effect of ageing and oxidative stress on antioxidant enzyme activity in different regions of the rat kidney. Mol Cell Biochem 2015; 408:253-60. [PMID: 26169983 DOI: 10.1007/s11010-015-2503-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/04/2015] [Indexed: 11/24/2022]
Abstract
Oxidative stress has been implicated in ageing and the pathogenesis of chronic kidney disease. We examined levels of antioxidant enzymes glutathione peroxidase, glutathione reductase, glutathione S-transferase, catalase and superoxide dismutase as modulated by age and oxidative stress in different regions of the kidney. Antioxidant enzymes were examined in different regions of the kidney in male Wistar rats. Kidneys from rats of different ages (5, 12, 36 and 60 weeks) were dissected into cortex, outer medulla and inner medulla. Tissues were incubated for 30 min with or without 0.2 mM H2O2 to induce oxidative stress. Antioxidant enzyme activities progressively decreased with age under both control and stress conditions (P < 0.05) after peaking at 12 weeks. Antioxidant enzyme activities were greater in the cortex (P < 0.05) by comparison with the outer and inner medulla, respectively.
Collapse
Affiliation(s)
- Noor Riyadh Thiab
- Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, NSW, 2351, Australia
| | - Nicola King
- School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, PL4 8AA, UK
| | - Graham L Jones
- Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
48
|
Lin CS, Lee SH, Huang HS, Chen YS, Ma MC. H2O2 generated by NADPH oxidase 4 contributes to transient receptor potential vanilloid 1 channel-mediated mechanosensation in the rat kidney. Am J Physiol Renal Physiol 2015; 309:F369-76. [PMID: 26136558 DOI: 10.1152/ajprenal.00462.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/17/2015] [Indexed: 11/22/2022] Open
Abstract
The presence of NADPH oxidase (Nox) in the kidney, especially Nox4, results in H2O2 production, which regulates Na(+) excretion and urine formation. Redox-sensitive transient receptor potential vanilloid 1 channels (TRPV1s) are distributed in mechanosensory fibers of the renal pelvis and monitor changes in intrapelvic pressure (IPP) during urine formation. The present study tested whether H2O2 derived from Nox4 affects TRPV1 function in renal sensory responses. Perfusion of H2O2 into the renal pelvis dose dependently increased afferent renal nerve activity and substance P (SP) release. These responses were attenuated by cotreatment with catalase or TRPV1 blockers. In single unit recordings, H2O2 activated afferent renal nerve activity in response to rising IPP but not high salt. Western blots revealed that Nox2 (gp91(phox)) and Nox4 are both present in the rat kidney, but Nox4 is abundant in the renal pelvis and originates from dorsal root ganglia. This distribution was associated with expression of the Nox4 regulators p22(phox) and polymerase δ-interacting protein 2. Coimmunoprecipitation experiments showed that IPP increases polymerase δ-interacting protein 2 association with Nox4 or p22(phox) in the renal pelvis. Interestingly, immunofluorescence labeling demonstrated that Nox4 colocalizes with TRPV1 in sensory fibers of the renal pelvis, indicating that H2O2 generated from Nox4 may affect TRPV1 activity. Stepwise increases in IPP and saline loading resulted in H2O2 and SP release, sensory activation, diuresis, and natriuresis. These effects, however, were remarkably attenuated by Nox inhibition. Overall, these results suggest that Nox4-positive fibers liberate H2O2 after mechanostimulation, thereby contributing to a renal sensory nerve-mediated diuretic/natriuretic response.
Collapse
Affiliation(s)
- Chian-Shiung Lin
- Department of Surgery, Liou-Ying Hospital, Chi-Mei Medical Center, Tainan Hsien, Taiwan
| | - Shang-Hsing Lee
- Department of Urology, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Ho-Shiang Huang
- Department of Urology, National Cheng Kung University, Tainan, Taiwan
| | - Yih-Sharng Chen
- Department of Cardiovascular Surgery, National Taiwan University Hospital, Taipei, Taiwan; and
| | - Ming-Chieh Ma
- School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| |
Collapse
|
49
|
Narne P, Ponnaluri KC, Siraj M, Ishaq M. Polymorphisms in oxidative stress pathway genes and risk of diabetic nephropathy in South Indian type 2 diabetic patients. Nephrology (Carlton) 2015; 19:623-9. [PMID: 25041504 DOI: 10.1111/nep.12293] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 11/29/2022]
Abstract
AIM Diabetic nephropathy (DN), a common microvascular complication of type 2 diabetes mellitus (T2DM) is polygenic, with a vast array of genes contributing to disease susceptibility. Accordingly, we explored the association between DN and six polymorphisms in oxidative stress related genes, namely eNOS, p22phox subunit of NAD(P)H oxidase, PARP-1 and XRCC1 in South Indian T2DM subjects. METHODS The study included 155 T2DM subjects with DN and 162 T2DM patients with no evidence of DN. The selected polymorphisms were genotyped by polymerase chain reaction and Taqman allele discrimination assay. RESULTS No significant difference was observed in the genotype and allele distribution of eNOS -786T > C, intron 4a4b, p22phox 242C > T and XRCC1 Arg399Gln polymorphisms between T2DM groups with and without DN. Contrastingly, there appeared to be a significant association of eNOS 894G > T and PARP-1 Val762Ala polymorphisms with DN wherein, the presence of 894T allele was associated with an enhanced risk for DN [P = 0.005; OR = 1.78 (1.17-2.7)], while the 762Ala allele seemed to confer significant protection against DN [P = 0.02; OR = 0.59 (0.37-0.92)]. Multiple logistic regression analysis revealed a significant and independent association of eNOS 894G > T, PARP-1 Val762Ala polymorphisms and hypertension with DN in T2DM individuals. CONCLUSIONS eNOS 894G > T and PARP-1 Val762Ala polymorphisms appeared to associate significantly with DN, with the former contributing to an enhanced risk and the latter to a reduced susceptibility to DN in South Indian T2DM individuals.
Collapse
Affiliation(s)
- Parimala Narne
- Department of Genetics, Osmania University, Hyderabad, India
| | | | | | | |
Collapse
|
50
|
Salehpour F, Ghanian Z, Yang C, Zheleznova NN, Kurth T, Dash RK, Cowley AW, Ranji M. Effects of p67phox on the mitochondrial oxidative state in the kidney of Dahl salt-sensitive rats: optical fluorescence 3-D cryoimaging. Am J Physiol Renal Physiol 2015; 309:F377-82. [PMID: 26062875 DOI: 10.1152/ajprenal.00098.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
The goal of the present study was to quantify and correlate the contribution of the cytosolic p67(phox) subunit of NADPH oxidase 2 to mitochondrial oxidative stress in the kidneys of the Dahl salt-sensitive (SS) hypertensive rat. Whole kidney redox states were uniquely assessed using a custom-designed optical fluorescence three-dimensional cryoimager to acquire multichannel signals of the intrinsic fluorophores NADH and FAD. SS rats were compared with SS rats in which the cytosolic subunit p67(phox) was rendered functionally inactive by zinc finger nuclease mutation of the gene (SS(p67phox)-null rats). Kidneys of SS rats fed a 0.4% NaCl diet exhibited significantly (P = 0.023) lower tissue redox ratio (NADH/FAD; 1.42 ± 0.06, n = 5) than SS(p67phox)-null rats (1.64 ± 0.07, n = 5), indicating reduced levels of mitochondrial electron transport chain metabolic activity and enhanced oxidative stress in SS rats. When fed a 4.0% salt diet for 21 days, both strains exhibited significantly lower tissue redox ratios (P < 0.001; SS rats: 1.03 ± 0.05, n = 9, vs. SS(p67phox)-null rats: 1.46 ± 0.04, n = 7) than when fed a 0.4% salt, but the ratio was still significantly higher in SS(p67phox) rats at the same salt level as SS rats. These results are consistent with results from previous studies that found elevated medullary interstitial fluid concentrations of superoxide and H2O2 in the medulla of SS rats. We conclude that the p67(phox) subunit of NADPH oxidase 2 plays an important role in the excess production of ROS from mitochondria in the renal medulla of the SS rat.
Collapse
Affiliation(s)
- F Salehpour
- Biophotonics Lab, University of Wisconsin, Milwaukee, Wisconsin; and
| | - Z Ghanian
- Biophotonics Lab, University of Wisconsin, Milwaukee, Wisconsin; and
| | - C Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - N N Zheleznova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - T Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - R K Dash
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - M Ranji
- Biophotonics Lab, University of Wisconsin, Milwaukee, Wisconsin; and
| |
Collapse
|