1
|
Du YX, Li X, Ji SW, Niu N. Hypertension toxicity of VEGFR-TKIs in cancer treatment: incidence, mechanisms, and management strategies. Arch Toxicol 2025; 99:67-81. [PMID: 39347999 DOI: 10.1007/s00204-024-03874-4] [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] [Received: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) are a class of targeted anticancer agents that include pazopanib, sunitinib, axitinib, and others. Currently, VEGFR-TKIs are widely used in the clinical treatment of various tumors, which can prolong patients' survival and even cure tumors. However, the use of VEGFR-TKIs is frequently associated with the occurrence of cardiovascular adverse events, with hypertension being the most prevalent. Hypertension and its complications can significantly impact the prognosis of patients, potentially jeopardizing their lives and resulting in the reduction or even cessation of treatment in severe cases. This review addresses the incidence of hypertension due to VEGFR-TKIs, mechanisms of toxicity, management strategies, and future research directions. In addition, hypertension due to VEGFR-TKIs may be associated with salt sensitivity, and possible mechanisms of hypertensive side effects are vasodilator imbalance, decreased capillary density, renal injury, impaired endothelial function due to oxidative stress, decreased lymphatic vascular density, and "off-target effect". A comprehensive understanding of hypertension toxicity due to cancer treatment with VEGFR-TKIs, can enhance clinical practice, thereby improving the prognostic outcomes of VEGFR-TKIs in oncology patients.
Collapse
Affiliation(s)
- Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Si-Wen Ji
- Office of Academic Affairs, North Sichuan Medical College, Nanchong, 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
2
|
Yang C, Isaeva E, Shimada S, Kurth T, Stumpf M, Zheleznova NN, Staruschenko A, Dash RK, Cowley AW. Inhibition of mTORC2 promotes natriuresis in Dahl salt-sensitive rats via the decrease of NCC and ENaC activity. Am J Physiol Renal Physiol 2024; 327:F435-F449. [PMID: 38779754 PMCID: PMC11460535 DOI: 10.1152/ajprenal.00403.2023] [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] [Received: 12/18/2023] [Revised: 03/27/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024] Open
Abstract
We have previously observed that prolonged administration of rapamycin, an inhibitor targeting the mammalian target of rapamycin complex (mTORC)1, partially reduced hypertension and alleviated kidney inflammation in Dahl salt-sensitive (SS) rats. In contrast, treatment with PP242, an inhibitor affecting both mTORC1/mTORC2, not only completely prevented hypertension but also provided substantial protection against kidney injury. Notably, PP242 exhibited potent natriuretic effects that were not evident with rapamycin. The primary objective of this study was to pinpoint the specific tubular sites responsible for the natriuretic effect of PP242 in SS rats subjected to either 0.4% NaCl (normal salt) or 4.0% NaCl (high salt) diet. Acute effects of PP242 on natriuretic, diuretic, and kaliuretic responses were determined in unanesthetized SS rats utilizing benzamil, furosemide, or hydrochlorothiazide [inhibitors of epithelial Na+ channel (ENaC), Na-K-2Cl cotransporter (NKCC2), or Na-Cl cotransporter (NCC), respectively] either administered alone or in combination. The findings indicate that the natriuretic effects of PP242 in SS rats stem predominantly from the inhibition of NCC and a reduction of ENaC open probability. Molecular analysis revealed that mTORC2 regulates NCC activity through protein phosphorylation and ENaC activity through proteolytic cleavage in vivo. Evidence also indicated that PP242 also prevents the loss of K+ associated with the inhibition of NCC. These findings suggest that PP242 may represent an improved therapeutic approach for antihypertensive intervention, potentially controlling blood pressure and mitigating kidney injury in salt-sensitive human subjects.NEW & NOTEWORTHY This study explored mechanisms underlying the natriuretic effects of mammalian target of rapamycin protein complex 2 inhibition using PP242 and revealed both epithelial Na+ channel and Na-Cl cotransporter in the distal tubular segments were potentially inhibited. These observations, with prior lab evidence, indicate that PP242 prevents hypertension via its potent inhibitory effects on these specific sodium transporters and by reducing renal immune responses. This dual action, coupled with potassium sparing effects, suggests an improved approach for managing hypertension and associated kidney damage.
Collapse
Affiliation(s)
- Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Elena Isaeva
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Megan Stumpf
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Nadezhda N Zheleznova
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Ranjan K Dash
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
3
|
Arkhipov SN, Liao TD, Potter DL, Bobbitt KR, Ivanov V, Ortiz PA, Pavlov TS. Dissociation of Hypertension and Renal Damage After Cessation of High-Salt Diet in Dahl Rats. Hypertension 2024; 81:1345-1355. [PMID: 38618734 PMCID: PMC11096017 DOI: 10.1161/hypertensionaha.123.21887] [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] [Received: 08/04/2023] [Accepted: 03/29/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Every year, thousands of patients with hypertension reduce salt consumption in an effort to control their blood pressure. However, hypertension has a self-sustaining character in a significant part of the population. We hypothesized that chronic hypertension leads to irreversible renal damage that remains after removing the trigger, causing an elevation of the initial blood pressure. METHODS Dahl salt-sensitive rat model was used for chronic, continuous observation of blood pressure. Rats were fed a high salt diet to induce hypertension, and then the diet was switched back to normal sodium content. RESULTS We found that developed hypertension was irreversible by salt cessation: after a short period of reduction, blood pressure grew even higher than in the high-salt phase. Notably, the self-sustaining phase of hypertension was sensitive to benzamil treatment due to sustaining epithelial sodium channel hyperactivity, as shown with patch-clamp analysis. Glomerular damage and proteinuria were also irreversible. In contrast, some mechanisms, contributing to the development of salt-sensitive hypertension, normalized after salt restriction. Thus, flow cytometry demonstrated that dietary salt reduction in hypertensive animals decreased the number of total CD45+, CD3+CD4+, and CD3+CD8+ cells in renal tissues. Also, we found tubular recovery and improvement of glomerular filtration rate in the postsalt period versus a high-salt diet. CONCLUSIONS Based on earlier publications and current data, poor response to salt restriction is due to the differential contribution of the factors recognized in the developmental phase of hypertension. We suggest that proteinuria or electrolyte transport can be prioritized over therapeutic targets of inflammatory response.
Collapse
Affiliation(s)
- Sergey N. Arkhipov
- Division of Hypertension and Vascular Research, Henry Ford Health
- Department of Physiology, Wayne State University
| | - Tang-Dong Liao
- Division of Hypertension and Vascular Research, Henry Ford Health
| | - D'Anna L. Potter
- Division of Hypertension and Vascular Research, Henry Ford Health
| | | | - Veniamin Ivanov
- Division of Hypertension and Vascular Research, Henry Ford Health
| | - Pablo A. Ortiz
- Division of Hypertension and Vascular Research, Henry Ford Health
- Department of Physiology, Wayne State University
| | - Tengis S. Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Health
- Department of Physiology, Wayne State University
| |
Collapse
|
4
|
Brobak KM, Melsom T, Eriksen BO, Høieggen A, Norvik JV, Solbu MD. The Association between Urinary Sodium-Potassium Ratio, Kidney Function, and Blood Pressure in a Cohort from the General Population. Kidney Blood Press Res 2024; 49:184-195. [PMID: 38382490 DOI: 10.1159/000535977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/07/2023] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Subclinical kidney dysfunction may contribute to salt-sensitive hypertension. We assessed the association between the urinary sodium-potassium ratio (Na/K ratio) and blood pressure (BP) in a general population cohort without diabetes, chronic kidney disease, cardiovascular disease, or treated hypertension. We investigated whether any such association was mediated by the kidney function markers measured glomerular filtration rate (mGFR), urinary albumin-creatinine ratio (ACR), and urinary epidermal growth factor-creatinine ratio (EGF-Cr). METHODS The Tromsø Study is a population-based study of inhabitants of the municipality of Tromsø, Northern Norway. Participants aged 50-62 years, without diabetes, chronic kidney disease, or cardiovascular disease, were invited to the substudy Renal Iohexol Clearance Survey in Tromsø 6 (RENIS-T6; 2007-09). For the present study, we excluded participants reporting the use of 1 or more antihypertensive agents, leaving 1,311 RENIS-T6 participants for a cross-sectional analysis. We measured office BP, 24-h ambulatory blood pressure (ABP), and mGFR using iohexol clearance. Na/K ratio, ACR, and EGF-Cr were measured in morning urine samples. RESULTS Urinary Na/K ratio was significantly associated with systolic office BP and ABP independently of cardiovascular risk factors and kidney function markers. A one-standard deviation unit increase in the Na/K ratio was associated with increased systolic ABP by 1.0 (0.3-1.6) mm Hg. Urinary Na/K ratio showed a stronger association with office BP than ABP. EGF-Cr, ACR, and mGFR did not mediate the relationship between urinary Na/K ratio and systolic BP. CONCLUSIONS In a representative sample of the middle-aged North-European population without diabetes, chronic kidney disease, cardiovascular disease, or treated hypertension, there was a consistent association between urinary Na/K ratio and BP. The association with BP was not mediated through kidney function measures, suggesting a relationship between a diet with high sodium and low potassium and higher BP regardless of kidney function.
Collapse
Affiliation(s)
- Karl Marius Brobak
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Toralf Melsom
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bjørn Odvar Eriksen
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Aud Høieggen
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jon Viljar Norvik
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Marit Dahl Solbu
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
5
|
Iwata Y, Deng Q, Kakizoe Y, Nakagawa T, Miyasato Y, Nakagawa M, Nishiguchi K, Nagayoshi Y, Narita Y, Izumi Y, Kuwabara T, Adachi M, Mukoyama M. A Serine Protease Inhibitor, Camostat Mesilate, Suppresses Urinary Plasmin Activity and Alleviates Hypertension and Podocyte Injury in Dahl Salt-Sensitive Rats. Int J Mol Sci 2023; 24:15743. [PMID: 37958726 PMCID: PMC10650472 DOI: 10.3390/ijms242115743] [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] [Received: 10/07/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
In proteinuric renal diseases, the serine protease (SP) plasmin activates the epithelial sodium channel (ENaC) by cleaving its γ subunit. We previously demonstrated that a high-salt (HS) diet provoked hypertension and proteinuria in Dahl salt-sensitive (DS) rats, accompanied by γENaC activation, which were attenuated by camostat mesilate (CM), an SP inhibitor. However, the effects of CM on plasmin activity in DS rats remain unclear. In this study, we investigated the effects of CM on plasmin activity, ENaC activation, and podocyte injury in DS rats. The DS rats were divided into the control diet, HS diet (8.0% NaCl), and HS+CM diet (0.1% CM) groups. After weekly blood pressure measurement and 24-h urine collection, the rats were sacrificed at 5 weeks. The HS group exhibited hypertension, massive proteinuria, increased urinary plasmin, and γENaC activation; CM treatment suppressed these changes. CM prevented plasmin(ogen) attachment to podocytes and mitigated podocyte injury by reducing the number of apoptotic glomerular cells, inhibiting protease-activated receptor-1 activation, and suppressing inflammatory and fibrotic cytokine expression. Our findings highlight the detrimental role of urinary plasmin in the pathogenesis of salt-sensitive hypertension and glomerular injury. Targeting plasmin with SP inhibitors, such as CM, may be a promising therapeutic approach for these conditions.
Collapse
Affiliation(s)
- Yasunobu Iwata
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Qinyuan Deng
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Yutaka Kakizoe
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
- Comprehensive Clinical Education, Training and Development Center, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Terumasa Nakagawa
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Yoshikazu Miyasato
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Miyuki Nakagawa
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Kayo Nishiguchi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Yu Nagayoshi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Yuki Narita
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Masataka Adachi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto 860-8556, Japan
- Comprehensive Clinical Education, Training and Development Center, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| |
Collapse
|
6
|
Sen T, Ju W, Nair V, Ladd P, Menon R, Otto EA, Pyle L, Vigers T, Nelson RG, Arnott C, Neal B, Hansen MK, Kretzler M, Bjornstad P, Heerspink HJL. Sodium glucose co-transporter 2 inhibition increases epidermal growth factor expression and improves outcomes in patients with type 2 diabetes. Kidney Int 2023; 104:828-839. [PMID: 37543256 DOI: 10.1016/j.kint.2023.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/30/2023] [Accepted: 07/21/2023] [Indexed: 08/07/2023]
Abstract
Underlying molecular mechanisms of the kidney protective effects of sodium glucose co-transporter 2 (SGLT2) inhibitors are not fully elucidated. Therefore, we studied the association between urinary epidermal growth factor (uEGF), a mitogenic factor involved in kidney repair, and kidney outcomes in patients with type 2 diabetes (T2D). The underlying molecular mechanisms of the SGLT2 inhibitor canagliflozin on EGF using single-cell RNA sequencing from kidney tissue were examined. Urinary EGF-to-creatinine ratio (uEGF/Cr) was measured in 3521 CANagliflozin cardioVascular Assessment Study (CANVAS) participants at baseline and week 52. Associations of uEGF/Cr with kidney outcome were assessed using multivariable-adjusted Cox regression models. Single-cell RNA sequencing was performed using protocol kidney biopsy tissue from ten young patients with T2D on SGLT2i, six patients with T2D on standard care only, and six healthy controls (HCs). In CANVAS, each doubling in baseline uEGF/Cr was associated with a 12% (95% confidence interval 1-22) decreased risk of kidney outcome. uEGF/Cr decreased after 52 weeks with placebo and remained stable with canagliflozin (between-group difference +7.3% (2.0-12.8). In young persons with T2D, EGF mRNA was primarily expressed in the thick ascending loop of Henle. Expression in biopsies from T2D without SGLT2i was significantly lower compared to HCs, whereas treatment with SGLT2i increased EGF levels closer to the healthy state. In young persons with T2D without SGLT2i, endothelin-1 emerged as a key regulator of the EGF co-expression network. SGLT2i treatment was associated with a shift towards normal EGF expression. Thus, decreased uEGF represents increased risk of kidney disease progression in patients with T2D. Canagliflozin increased kidney tissue expression of EGF and was associated with a downstream signaling cascade linked to tubular repair and reversal of tubular injury.
Collapse
Affiliation(s)
- Taha Sen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patricia Ladd
- Department of Radiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Laura Pyle
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA; Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Tim Vigers
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA; Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Clare Arnott
- The George Institute for Global Health, University New South Wales (UNSW), Sydney, New South Wales, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Bruce Neal
- The George Institute for Global Health, University New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Michael K Hansen
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA.
| | - Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA; Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA.
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands; The George Institute for Global Health, University New South Wales (UNSW), Sydney, New South Wales, Australia.
| |
Collapse
|
7
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic Responses of Normal Rat Kidneys to a High Salt Intake. FUNCTION 2023; 4:zqad031. [PMID: 37575482 PMCID: PMC10413938 DOI: 10.1093/function/zqad031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 08/15/2023] Open
Abstract
In this study, novel methods were developed, which allowed continuous (24/7) measurement of arterial blood pressure and renal blood flow in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O2 and metabolites. Specifically, the study determined the effects of a high salt (HS; 4.0% NaCl) diet upon whole kidney O2 consumption and arterial and renal venous plasma metabolomic profiles of normal Sprague-Dawley rats. A separate group of rats was studied to determine changes in the cortex and outer medulla tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to 4.0% NaCl diet. In addition, targeted mRNA expression analysis of cortical segments was performed. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. A novel finding was the increased expression of glycolysis-related genes in Cx and isolated proximal tubular segments in response to an HS diet, consistent with increased release of pyruvate and lactate from the kidney to the renal venous blood. Data suggests that aerobic glycolysis (eg, Warburg effect) may contribute to energy production under these circumstances. The study provides evidence that kidney metabolism responds to an HS diet enabling enhanced energy production while protecting from oxidative stress and injury. Metabolomic and transcriptomic analysis of kidneys of Sprague-Dawley rats fed a high salt diet.
Collapse
Affiliation(s)
- Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian R Hoffmann
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Andrew S Greene
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K Dash
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI 53226, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
8
|
Gao ZX, Wei QC, Shu TT, Li ST, Zhou R, Li MY, Mao ZH, Liu DW, Liu ZS, Wu P. Kir4.1 deletion prevents salt-sensitive hypertension in early streptozotocin-induced diabetic mice via Na + -Cl - cotransporter in the distal convoluted tubule. J Hypertens 2023; 41:958-970. [PMID: 37016934 DOI: 10.1097/hjh.0000000000003419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
OBJECTIVES Functional impairment of renal sodium handling and blood pressure (BP) homeostasis is an early characteristic manifestation of type 1 diabetes. However, the underlying mechanisms remain unclear. METHODS Metabolic cages, radio-telemetry, immunoblotting, and electrophysiology were utilized to examine effects of high salt (8% NaCl, HS) intake on Na + /K + balance, BP, Na + -Cl - cotransporter (NCC) function, and basolateral K + channel activity in the distal convoluted tubule (DCT) under diabetic conditions. RESULTS Improper Na + balance, hypernatremia, and a mild but significant increase in BP were found in streptozotocin (STZ)-induced diabetic mice in response to HS intake for 7 days. Compared to the vehicle, STZ mice showed increased Kir4.1 expression and activity in the DCT, a more negative membrane potential, higher NCC abundance, and enhanced hydrochlorothiazide-induced natriuretic effect. However, HS had no significant effect on basolateral Kir4.1 expression/activity and DCT membrane potential, or NCC activity under diabetic conditions, despite a downregulation in phosphorylated NCC abundance. In contrast, HS significantly downregulated the expression of Na + -H + exchanger 3 (NHE3) and cleaved epithelial sodium channel-γ in STZ mice, despite an increase in NHE3 abundance after STZ treatment. Kir4.1 deletion largely abolished STZ-induced upregulation of NCC expression and prevented BP elevation during HS intake. Interestingly, HS causes severe hypokalemia in STZ-treated kidney-specific Kir4.1 knockout (Ks-Kir4.1 KO) mice and lead to death within a few days, which could be attributed to a higher circulating aldosterone level. CONCLUSIONS We concluded that Kir4.1 is required for upregulating NCC activity and may be essential for developing salt-sensitive hypertension in early STZ-induced diabetes.
Collapse
Affiliation(s)
- Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Qi-Chao Wei
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Ting-Ting Shu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shu-Ting Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Rui Zhou
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Ming-Yan Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University
- Institute of Nephrology, Zhengzhou University
- Henan Province Clinical Research Center for Kidney Disease
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
9
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic responses of normal rat kidneys to a high salt intake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524636. [PMID: 36711564 PMCID: PMC9882299 DOI: 10.1101/2023.01.18.524636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the present study, novel methods were developed which allowed continuous (24/7) measurement of blood pressure (BP) and renal blood flow (RBF) in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O 2 and metabolites. The study determined the effects of a high salt (HS) diet upon whole kidney O 2 consumption and the metabolomic profiles of normal Sprague Dawley (SD) rats. A separate group of rats was studied to determine changes in the cortex (Cx) and outer medulla (OM) tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to a 4.0% NaCl diet. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O 2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. Increased glycolysis was evident with the elevation of mRNA expression encoding key glycolytic enzymes and release of pyruvate and lactate from the kidney in the renal venous blood. Glycolytic production of NADH is used in either the production of lactate or oxidized via the malate aspartate shuttle. Aerobic glycolysis (e.g., Warburg-effect) may account for the needed increase in cellular energy. The study provides evidence that kidney metabolism responds to a HS diet enabling enhanced energy production while protecting from oxidate stress and injury.
Collapse
|
10
|
Renal lysophospholipase A1 contributes to Enterococcus faecalis-induced hypertension by enhancing sodium reabsorption. iScience 2022; 25:105403. [DOI: 10.1016/j.isci.2022.105403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/23/2022] [Accepted: 10/16/2022] [Indexed: 11/17/2022] Open
|
11
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
12
|
Rinschen MM, Palygin O, El-Meanawy A, Domingo-Almenara X, Palermo A, Dissanayake LV, Golosova D, Schafroth MA, Guijas C, Demir F, Jaegers J, Gliozzi ML, Xue J, Hoehne M, Benzing T, Kok BP, Saez E, Bleich M, Himmerkus N, Weisz OA, Cravatt BF, Krüger M, Benton HP, Siuzdak G, Staruschenko A. Accelerated lysine metabolism conveys kidney protection in salt-sensitive hypertension. Nat Commun 2022; 13:4099. [PMID: 35835746 PMCID: PMC9283537 DOI: 10.1038/s41467-022-31670-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/27/2022] [Indexed: 01/07/2023] Open
Abstract
Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.
Collapse
Affiliation(s)
- Markus M Rinschen
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- III. Medical Clinic, University Hospital Hamburg Eppendorf, Hamburg, Germany.
- AIAS, Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark.
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xavier Domingo-Almenara
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Omics Sciences Unit, EURECAT, Technology Centre of Catalonia, Reus, Catalonia, Spain
| | - Amelia Palermo
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Fatih Demir
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Megan L Gliozzi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jingchuan Xue
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Martin Hoehne
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Thomas Benzing
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Bernard P Kok
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Markus Bleich
- Institute of Physiology, University Kiel, Kiel, Germany
| | | | - Ora A Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | | | - Marcus Krüger
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - H Paul Benton
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
13
|
Shimada S, Yang C, Kumar V, Mattson DL, Cowley AW. Acute Increase of Renal Perfusion Pressure Causes Rapid Activation of mTORC1 (Mechanistic Target Of Rapamycin Complex 1) and Leukocyte Infiltration. Hypertension 2022; 79:1180-1189. [PMID: 35291809 PMCID: PMC9098670 DOI: 10.1161/hypertensionaha.121.18643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/25/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The present study in Sprague-Dawley rats determined the effects of a rapid rise of renal perfusion pressure (RPP) upon the activation of mTOR (mechanistic target of rapamycin), and the effects upon the infiltration of CD68-positive macrophages/monocytes and CD3-positive T lymphocytes into the kidneys. METHODS RPP was elevated by 40 mm Hg for 30 minutes in male Sprague-Dawley rats while measuring renal blood flow and urine flow rate. Sham rats were studied in the same way, but RPP was not changed. Since initial studies found that the acute increase of RPP resulted in activation of mTORC1 (phosphorylation of S6S235/236), the effects of inhibition of mTORC1 with rapamycin pretreatment were then determined. RESULTS It was found that a 30-minute increase of RPP (≈40 mm Hg) resulted in an 8-fold increase of renal sodium excretion which was blunted by rapamycin treatment. Renal blood flow was not affected by the elevation of RPP. Activation of mTORC1 was observed. Significant increases in CD68-positive macrophages were found in both the cortex (intraglomerular and periglomerular regions) and in the outer medullary interstitial regions of the kidney and prevented by rapamycin treatment. Increases in CD3-positive T lymphocytes were observed exclusively in the periglomerular regions and prevented by rapamycin treatment. Upregulation of several proinflammatory markers was observed. CONCLUSIONS We conclude that elevation of RPP rapidly activates mTORC1 resulting in infiltration of immune cells into the kidney.
Collapse
Affiliation(s)
- Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee (S.S., C.Y., V.K., D.L.M., A.W.C.)
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee (S.S., C.Y., V.K., D.L.M., A.W.C.)
| | - Vikash Kumar
- Department of Physiology, Medical College of Wisconsin, Milwaukee (S.S., C.Y., V.K., D.L.M., A.W.C.)
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee (S.S., C.Y., V.K., D.L.M., A.W.C.)
- Now with: Department of Physiology, Medical College of Georgia at Augusta University (D.L.M.)
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee (S.S., C.Y., V.K., D.L.M., A.W.C.)
| |
Collapse
|
14
|
Kravtsova O, Bohovyk R, Levchenko V, Palygin O, Klemens CA, Rieg T, Staruschenko A. SGLT2 inhibition effect on salt-induced hypertension, RAAS, and Na + transport in Dahl SS rats. Am J Physiol Renal Physiol 2022; 322:F692-F707. [PMID: 35466690 PMCID: PMC9142161 DOI: 10.1152/ajprenal.00053.2022] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022] Open
Abstract
Na+-glucose cotransporter-2 (SGLT2) inhibitors are the new mainstay of treatment for diabetes mellitus and cardiovascular diseases. Despite the remarkable benefits, the molecular mechanisms mediating the effects of SGLT2 inhibitors on water and electrolyte balance are incompletely understood. The goal of this study was to determine whether SGLT2 inhibition alters blood pressure and kidney function via affecting the renin-angiotensin-aldosterone system (RAAS) and Na+ channels/transporters along the nephron in Dahl salt-sensitive rats, a model of salt-induced hypertension. Administration of dapagliflozin (Dapa) at 2 mg/kg/day via drinking water for 3 wk blunted the development of salt-induced hypertension as evidenced by lower blood pressure and a left shift of the pressure natriuresis curve. Urinary flow rate, glucose excretion, and Na+- and Cl--to-creatinine ratios increased in Dapa-treated compared with vehicle-treated rats. To define the contribution of the RAAS, we measured various hormones. Despite apparent effects on Na+- and Cl--to-creatinine ratios, Dapa treatment did not affect RAAS metabolites. Subsequently, we assessed the effects of Dapa on renal Na+ channels and transporters using RT-PCR, Western blot analysis, and patch clamp. Neither mRNA nor protein expression levels of renal transporters (SGLT2, Na+/H+ exchanger isoform 3, Na+-K+-2Cl- cotransporter 2, Na+-Cl- cotransporter, and α-, β-, and γ-epithelial Na+ channel subunits) changed significantly between groups. Furthermore, electrophysiological experiments did not reveal any difference in Dapa treatment on the conductance and activity of epithelial Na+ channels. Our data suggest that SGLT2 inhibition in a nondiabetic model of salt-sensitive hypertension blunts the development of salt-induced hypertension by causing glucosuria and natriuresis without changes in the RAAS or the expression or activity of the main Na+ channels and transporters.NEW & NOTEWORTHY The present study indicates that Na+-glucose cotransporter-2 (SGLT2) inhibition in a nondiabetic model of salt-sensitive hypertension blunts the development and magnitude of salt-induced hypertension. Chronic inhibition of SGLT2 increases glucose and Na+ excretion without secondary effects on the expression and function of other Na+ transporters and channels along the nephron and hormone levels in the renin-angiotensin-aldosterone system. These data provide novel insights into the effects of SGLT2 inhibitors and their potential use in hypertension.
Collapse
Affiliation(s)
- Olha Kravtsova
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Ruslan Bohovyk
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Oleg Palygin
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Christine A Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- James A. Haley Veterans' Hospital, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- James A. Haley Veterans' Hospital, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
| |
Collapse
|
15
|
Zeid AM, Lamontagne JO, Zhang H, Marneros AG. Epidermal growth factor deficiency predisposes to progressive renal disease. FASEB J 2022; 36:e22286. [PMID: 35442545 DOI: 10.1096/fj.202101837r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/05/2022] [Accepted: 03/17/2022] [Indexed: 01/22/2023]
Abstract
Epidermal growth factor (EGF) is produced in the kidney by thick ascending limbs of the loop of Henle and by distal convoluted tubules (DCTs). Reduced urinary EGF levels have been associated with chronic kidney disease but it is not known whether physiological levels of EGF protect the kidney from progressive renal disease. Here, we show that EGF-deficient mice on a mixed genetic background had increased urinary microalbumin, and a subset of these mice developed severe progressive renal disease with azotemia that was not seen in WT or TGFα-deficient littermates with this mixed genetic background. These azotemic EGF-deficient mice developed crescentic glomerulonephritis linked to HB-EGF/EGFR hyperactivation in glomeruli, as well as attenuation of the proximal tubule brush border, distal convoluted tubule (DCT) dilatation, and kidney fibrosis associated with renal β-catenin/mTOR hyperactivation. The observation of these severe renal pathologies only in a subset of EGF-deficient mice suggests that independent segregation of strain-specific modifier alleles contributes to the severity of the renal abnormalities that only manifest when EGF is lacking. These findings link the lack of EGF to renal pathologies in the adult mammalian kidney, in support of a role of physiological levels of EGF for maintaining the function of glomeruli, proximal tubules, and DCTs. These observations suggest that diminished EGF levels predispose kidneys to progressive renal disease.
Collapse
Affiliation(s)
- Alia M Zeid
- Department of Dermatology, Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Joseph O Lamontagne
- Department of Dermatology, Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hui Zhang
- Department of Dermatology, Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Alexander G Marneros
- Department of Dermatology, Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
16
|
Mining Important Herb Combinations of Traditional Chinese Medicine against Hypertension Based on the Symptom-Herb Network Combined with Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5850899. [PMID: 35360657 PMCID: PMC8964163 DOI: 10.1155/2022/5850899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 11/18/2022]
Abstract
Although data mining methods are extensively used in the rule analysis of famous old traditional Chinese medicine (TCM) experts' prescriptions for the treatment of hypertension, most of them only mine the association between herbs and herbs, ignoring the importance of symptoms in the disease. This study collected 439 cases of hypertension treated by famous old TCM experts from the FangNet platform. Using the structure network algorithm, the symptom-herb network was constructed, which redefined the importance of herb in disease. Based on the network, 21 driver herbs, 76 herb pairs, and 41 symptom-herb associations were mined. Finally, the basic prescription composed of Gouteng (Uncariae Ramulus cum Uncis), Huanglian (Coptidis Rhizoma), Chuanxiong (Chuanxiong Rhizoma), Gegen (Puerariae Lobatae Radix), Danggui (Angelicae Sinensis Radix), and Huangqin (Scutellariae Radix) was found. These herbs are the most significant among all herbs, and they have a potential correlation with each other. To further verify the rationality of the data mining results, we adopted the network pharmacology method. Network pharmacological analysis shows that the five core targets in the basic prescription include IL6, VEGFA, TNF, TP53, and EGF, which link 10 significant active compounds and 7 important KEGG pathways. It was predicted that anti-inflammatory, antioxidant, vascular endothelial protection, emotion regulation, and ion channel intervention might be the main mechanisms of the basic prescription against hypertension. This study reveals the prescription rule of famous old TCM experts for treating hypertension from a new perspective, which provides a new approach to inherit the academic experience of famous old TCM experts and develop new drugs.
Collapse
|
17
|
Corrêa JWN, Boaro KR, Sene LB, Polidoro JZ, Salles TA, Martins FL, Bendhack LM, Girardi ACC. Antiproteinuric and Hyperkalemic Mechanisms Activated by Dual Versus Single Blockade of the RAS in Renovascular Hypertensive Rats. Front Physiol 2021; 12:656460. [PMID: 34177612 PMCID: PMC8221266 DOI: 10.3389/fphys.2021.656460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022] Open
Abstract
This study aimed to investigate the antiproteinuric and hyperkalemic mechanisms activated by dual renin-angiotensin system (RAS) blockade in renovascular hypertensive rats (2-kidney 1-clip model [2K-1C]). Six weeks after clipping the left renal artery or sham operation (2K), rats were treated with losartan, enalapril, or both drugs for two weeks. We found that 2K-1C rats displayed higher tail-cuff blood pressure (BP), increased non-clipped kidney Ang II concentration, and more pronounced urinary albumin excretion than 2K. BP was decreased by the treatment with either enalapril or losartan, and the combination of both drugs promoted an additional antihypertensive effect in 2K-1C rats. Renal Ang II content and albuminuria were reduced by either enalapril or losartan in monotherapy and restored to control levels by dual RAS blockade. Albuminuria in 2K-1C rats was accompanied by downregulation of the glomerular slit protein podocin, reduction of the endocytic receptors megalin and cubilin, and a marked decrease in the expression of the ClC-5 chloride channel, compared to 2K animals. Treatment with losartan and enalapril in monotherapy or combination increased the expression of podocin, cubilin, and ClC-5. However, only the combined therapy normalized podocin, cubilin, and ClC-5 protein abundance in the non-clipped kidney of 2K-1C rats. Renovascular hypertensive 2K-1C rats had a lower concentration of plasma potassium compared to 2K rats. Single RAS blockade normalized potassium plasma concentration, whereas 2K-1C rats treated with dual RAS blockade exhibited hyperkalemia. Hypokalemia in 2K-1C rats was accompanied by an increase in the cleaved activated forms of α-ENaC and γ-ENaC and the expression of β-ENaC. Combined RAS blockade but not monotherapy significantly reduced the expression of these ENaC subunits in 2K-1C rats. Indeed, double RAS blockade reduced the abundance of cleaved-α-ENaC to levels lower than those of 2K rats. Collectively, these results demonstrate that the antiproteinuric effect of dual RAS blockade in 2K-1C rats is associated with the restored abundance of podocin and cubilin, and ClC-5. Moreover, double RAS blockade-induced hyperkalemia may be due, at least partially, to an exaggerated downregulation of cleaved α-ENaC in the non-clipped kidney of renovascular hypertensive rats.
Collapse
Affiliation(s)
- José Wilson N Corrêa
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Amazonas, Manaus, Brazil
| | - Karoline R Boaro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Letícia B Sene
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Juliano Z Polidoro
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Thiago A Salles
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Flavia L Martins
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| | - Lusiane M Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor) University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
18
|
Abstract
The development of high blood pressure is influenced by genetic and environmental factors, with high salt intake being a known environmental contributor. Humans display a spectrum of sodium-sensitivity, with some individuals displaying a significant blood pressure rise in response to increased sodium intake while others experience almost no change. These differences are, in part, attributable to genetic variation in pathways involved in sodium handling and excretion. ENaC (epithelial sodium channel) is one of the key transporters responsible for the reabsorption of sodium in the distal nephron. This channel has an important role in the regulation of extracellular fluid volume and consequently blood pressure. Herein, we review the role of ENaC in the development of salt-sensitive hypertension, and present mechanistic insights into the regulation of ENaC activity and how it may accelerate sodium-induced damage and dysfunction. We discuss the traditional role of ENaC in renal sodium reabsorption and review work addressing ENaC expression and function in the brain, vasculature, and immune cells, and how this has expanded the implications for its role in the initiation and progression of salt-sensitive hypertension.
Collapse
Affiliation(s)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, and Department of Molecular Physiology and Biophysics Vanderbilt University, Nashville, TN (A.K.)
| | - Thomas R Kleyman
- From the Department of Medicine (S.M.M., T.R.K.), University of Pittsburgh, PA.,Department of Cell Biology (T.R.K.), University of Pittsburgh, PA.,Department of Pharmacology and Chemical Biology (T.R.K.), University of Pittsburgh, PA
| |
Collapse
|
19
|
Expression of Gallus Epidermal Growth Factor (gEGF) with Food-Grade Lactococcus lactis Expression System and Its Biological Effects on Broiler Chickens. Biomolecules 2021; 11:biom11010103. [PMID: 33466817 PMCID: PMC7829775 DOI: 10.3390/biom11010103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022] Open
Abstract
As a multifunctional polypeptide, epidermal growth factor (EGF) increases growth performance or enhances resistance to diseases in commercial broilers under adverse conditions. In this study, a recombinant Lactococcus lactis was established to produce the secretory form of bioactive gEGF. The results of in vitro testing showed that gEGF promoted the proliferation of chicken embryo fibroblast cells. A total of 63 5-day-old broiler chickens were evenly divided into three groups and treated with either M17 medium (the control group), supernatant of LL-pNZ8149 fermentation product (the P-LL group), or supernatant of LL-pNZ8149-gEGF fermentation product (the gEGF group). In two weeks, many measurements of growth, immunity and the intestines were significantly higher in the gEGF group than those in the control and the P-LL groups. Our study showed that the bioactive gEGF could be expressed with Lactococcus lactis expression system with the potential to enhance growth performance, immune function, and intestinal development in broiler chickens.
Collapse
|
20
|
Klemens CA, Chulkov EG, Wu J, Hye Khan MA, Levchenko V, Flister MJ, Imig JD, Kriegel AJ, Palygin O, Staruschenko A. Loss of Chloride Channel 6 (CLC-6) Affects Vascular Smooth Muscle Contractility and Arterial Stiffness via Alterations to Golgi Calcium Stores. Hypertension 2021; 77:582-593. [PMID: 33390052 DOI: 10.1161/hypertensionaha.120.16589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Genome-wide association studies have found a number of potential genes involved in blood pressure regulation; however, the functional role of many of these candidates has yet to be established. One such candidate gene is CLCN6, which encodes the transmembrane protein, chloride channel 6 (ClC-6). Although the CLCN6 locus has been widely associated with human blood pressure regulation, the mechanistic role of ClC-6 in blood pressure homeostasis at the molecular, cellular, and physiological levels is completely unknown. In this study, we demonstrate that rats with a functional knockout of ClC-6 on the Dahl Salt-Sensitive rat background (SS-Clcn6) have lower diastolic but not systolic blood pressures. The effect of diastolic blood pressure attenuation was independent of dietary salt exposure in knockout animals. Moreover, SS-Clcn6 rats are protected from hypertension-induced cardiac hypertrophy and arterial stiffening; however, they have impaired vasodilation and dysregulated intracellular calcium handling. ClC-6 is highly expressed in vascular smooth muscle cells where it is targeted to the Golgi apparatus. Using bilayer electrophysiology, we provide evidence that recombinant human ClC-6 protein can function as a channel. Last, we demonstrate that loss of ClC-6 function reduces Golgi calcium stores, which may play a previously unidentified role in vascular contraction and relaxation signaling in vascular smooth muscle cells. Collectively, these data indicate that ClC-6 may modulate blood pressure by regulating Golgi calcium reserves, which in turn contribute to vascular smooth muscle function.
Collapse
Affiliation(s)
- Christine A Klemens
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin.,Cardiovascular Center (C.A.K., J.W., J.D.I., O.P., A.S.), Medical College of Wisconsin
| | - Evgeny G Chulkov
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin.,Department of Cell Biology, Neurobiology and Anatomy (E.G.C.), Medical College of Wisconsin
| | - Jing Wu
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin.,Cardiovascular Center (C.A.K., J.W., J.D.I., O.P., A.S.), Medical College of Wisconsin
| | - Md Abdul Hye Khan
- Department of Pharmacology (M.A.H.K., J.D.I.), Medical College of Wisconsin
| | - Vladislav Levchenko
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin
| | - Michael J Flister
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin
| | - John D Imig
- Department of Pharmacology (M.A.H.K., J.D.I.), Medical College of Wisconsin.,Cardiovascular Center (C.A.K., J.W., J.D.I., O.P., A.S.), Medical College of Wisconsin
| | - Alison J Kriegel
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin
| | - Oleg Palygin
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin.,Cardiovascular Center (C.A.K., J.W., J.D.I., O.P., A.S.), Medical College of Wisconsin
| | - Alexander Staruschenko
- From the Department of Physiology (C.A.K., E.G.C., J.W., V.L., M.J.F., A.J.K., O.P., A.S.), Medical College of Wisconsin.,Cardiovascular Center (C.A.K., J.W., J.D.I., O.P., A.S.), Medical College of Wisconsin.,Clement J. Zablocki VA Medical Center, Milwaukee (A.S.)
| |
Collapse
|
21
|
Urinary proteomics reveals key markers of salt sensitivity in hypertensive patients during saline infusion. J Nephrol 2021; 34:739-751. [PMID: 33398797 DOI: 10.1007/s40620-020-00877-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypertension is a complex disease and is the major cause of cardiovascular complications. In the vast majority of individuals, the aetiology of elevated blood pressure (BP) cannot be determined, thus impairing optimized therapies and prognosis for individual patients. A more precise understanding of the molecular pathogenesis of hypertension remains a pressing priority for both basic and translational research. Here we investigated the effect of salt on naive hypertensive patients in order to better understand the salt intake-blood pressure relationship. METHODS Patients underwent an acute saline infusion and were defined as salt-sensitive or salt-resistant according to mean blood pressure changes. Urinary proteome changes during the salt load test were analysed by a label-free quantitative proteomics approach. RESULTS Our data show that salt-sensitive patients display equal sodium reabsorption as salt-resistant patients, as major sodium transporters show the same behaviour during the salt load. However, salt-sensitive patients regulate the renin angiotensin system (RAS) differently from salt-resistant patients, and upregulate proteins, as epidermal growth factor (EGF) and plasminogen activator, urokinase (PLAU), involved in the regulation of epithelial sodium channel ENaC activity. CONCLUSIONS Salt-sensitive and salt-resistant subjects have similar response to a saline/volume infusion as detected by urinary proteome. However, we identified glutamyl aminopeptidase (ENPEP), PLAU, EGF and Xaa-Pro aminopeptidase 2 precursor XPNPEP2 as key molecules of salt-sensitivity, through modulation of ENaC-dependent sodium reabsorption along the distal tubule.
Collapse
|
22
|
Pavlov TS, Palygin O, Isaeva E, Levchenko V, Khedr S, Blass G, Ilatovskaya DV, Cowley AW, Staruschenko A. NOX4-dependent regulation of ENaC in hypertension and diabetic kidney disease. FASEB J 2020; 34:13396-13408. [PMID: 32799394 DOI: 10.1096/fj.202000966rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022]
Abstract
NADPH oxidase 4 (NOX4) is the most abundant NOX isoform in the kidney; however, its importance for renal function has only recently emerged. The NOX4-dependent pathway regulates many factors essential for proper sodium handling in the distal nephron. However, the functional significance of this pathway in the control of sodium reabsorption during the initiation of chronic kidney disease is not established. The goal of this study was to test Nox4-dependent ENaC regulation in two models: SS hypertension and STZ-induced type 1 diabetes. First, we showed that genetic ablation of Nox4 in Dahl salt-sensitive (SS) rat attenuated a high-salt (HS)-induced increase in epithelial Na+ channel (ENaC) activity in the cortical collecting duct. We also found that H2 O2 upregulated ENaC activity, and H2 O2 production was reduced in both the renal cortex and medulla in SSNox4-/- rats fed an HS diet. Second, in the streptozotocin model of hyperglycemia-induced renal injury ENaC activity in hyperglycemic animals was elevated in SS but not SSNox4-/- rats. NaCl cotransporter (NCC) expression was increased compared to healthy controls, while expression values between SS and SSNox4-/- groups were similar. These data emphasize a critical contribution of the NOX4-mediated pathway in maladaptive upregulation of ENaC-mediated sodium reabsorption in the distal nephron in the conditions of HS- and hyperglycemia-induced kidney injury.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena Isaeva
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Gregory Blass
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Clement J. Zablocki VA Medical Center, Milwaukee, WI, USA
| |
Collapse
|
23
|
Romano R, Bucci C. Role of EGFR in the Nervous System. Cells 2020; 9:E1887. [PMID: 32806510 PMCID: PMC7464966 DOI: 10.3390/cells9081887] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is the first discovered member of the receptor tyrosine kinase superfamily and plays a fundamental role during embryogenesis and in adult tissues, being involved in growth, differentiation, maintenance and repair of various tissues and organs. The role of EGFR in the regulation of tissue development and homeostasis has been thoroughly investigated and it has also been demonstrated that EGFR is a driver of tumorigenesis. In the nervous system, other growth factors, and thus other receptors, are important for growth, differentiation and repair of the tissue, namely neurotrophins and neurotrophins receptors. For this reason, for a long time, the role of EGFR in the nervous system has been underestimated and poorly investigated. However, EGFR is expressed both in the central and peripheral nervous systems and it has been demonstrated to have specific important neurotrophic functions, in particular in the central nervous system. This review discusses the role of EGFR in regulating differentiation and functions of neurons and neuroglia. Furthermore, its involvement in regeneration after injury and in the onset of neurodegenerative diseases is examined.
Collapse
Affiliation(s)
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy;
| |
Collapse
|
24
|
Tu J, Zhang B, Fang G, Chang W, Zhao Y. Neddylation-mediated Nedd4-2 activation regulates ubiquitination modification of renal NBCe1. Exp Cell Res 2020; 390:111958. [DOI: 10.1016/j.yexcr.2020.111958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
|
25
|
Liu M, Deng M, Luo Q, Dou X, Jia Z. High-Salt Loading Downregulates Nrf2 Expression in a Sodium-Dependent Manner in Renal Collecting Duct Cells. Front Physiol 2020; 10:1565. [PMID: 32038274 PMCID: PMC6985211 DOI: 10.3389/fphys.2019.01565] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
Background High salt intake is associated with both oxidative stress and chronic kidney disease (CKD) progression. Nuclear factor E2-related factor 2 (Nrf2) is a transcriptional factor regulating the antioxidant and detoxifying genes to potently antagonize oxidative stress. This study examined the effect of high salt loading on the expression of Nrf2 in kidney. Methods Mice were treated with acute salt loading, and Nrf2 expression in the kidney was detected by Western blotting and immunostaining. Reactive oxygen species (ROS) levels in the kidney were measured using dihydroethidium (DHE) staining. In vitro, mpkCCD cells were cultured in high osmolality medium by adding sodium chloride (NaCl), sodium gluconate (Na-Glu), choline chloride (Choline-Cl), or mannitol. Then, Nrf2 and its target genes were measured. Results Nrf2 protein in renal cortex and medulla tissue lysates was significantly downregulated after acute salt loading. Immunofluorescence data showed that Nrf2 was mainly located in collecting duct principal cells evidenced by co-staining of Nrf2 with AQP2. Contrasting to the reduced Nrf2 expression, ROS levels in the kidney were significantly increased after salt loading. In vitro, the Nrf2 protein level was downregulated in mpkCCD cells after NaCl treatment for 24 h. Interestingly, sodium gluconate had a similar effect on downregulating Nrf2 expression as NaCl, whereas neither Choline-Cl nor mannitol changed Nrf2 expression. Meanwhile, the mRNA levels of Nrf2 target genes were downregulated by NaCl and/or sodium gluconate, while some of them were also regulated by Choline-Cl, indicating a more complex regulation of these genes under a high salt condition. Finally, we found that the downregulation of Nrf2 caused by NaCl was not affected by N-acetylcysteine (NAC), spironolactone, or NS-398, suggesting other mechanisms mediating Nrf2 downregulation caused by high salt challenge. Conclusion High salt downregulated Nrf2 mainly via a sodium-dependent manner in kidney collecting duct cells, which might contribute to the excessive renal oxidative stress and CKD progression.
Collapse
Affiliation(s)
- Mi Liu
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mokan Deng
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Qimei Luo
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Xianrui Dou
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Manis AD, Palygin O, Khedr S, Levchenko V, Hodges MR, Staruschenko A. Relationship between the renin-angiotensin-aldosterone system and renal Kir5.1 channels. Clin Sci (Lond) 2019; 133:2449-2461. [PMID: 31799617 PMCID: PMC8474144 DOI: 10.1042/cs20190876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/13/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
Kir5.1 (encoded by the Kcnj16 gene) is an inwardly rectifying K+ (Kir) channel highly expressed in the aldosterone-sensitive distal nephron of the kidney, where it forms a functional channel with Kir4.1. Kir4.1/Kir5.1 channels are responsible for setting the transepithelial voltage in the distal nephron and collecting ducts and are thereby major determinants of fluid and electrolyte distribution. These channels contribute to renal blood pressure control and have been implicated in salt-sensitive hypertension. However, mechanisms pertaining to the impact of K ir4.1/Kir5.1-mediated K+ transport on the renin-angiotensin-aldosterone system (RAAS) remain unclear. Herein, we utilized a knockout of Kcnj16 in the Dahl salt-sensitive rat (SSKcnj16-/-) to investigate the relationship between Kir5.1 and RAAS balance and function in the sensitivity of blood pressure to the dietary Na+/K+ ratio. The knockout of Kcnj16 caused substantial elevations in plasma RAAS hormones (aldosterone and angiotensin peptides) and altered the RAAS response to changing the dietary Na+/K+ ratio. Blocking aldosterone with spironolactone caused rapid mortality in SSKcnj16-/- rats. Supplementation of the diet with high K+ was protective against mortality resulting from aldosterone-mediated mechanisms. Captopril and losartan treatment had no effect on the survival of SSKcnj16-/- rats. However, neither of these drugs prevented mortality of SSKcnj16-/- rats when switched to high Na+ diet. These studies revealed that the knockout of Kcnj16 markedly altered RAAS regulation and function, suggesting Kir5.1 as a key regulator of the RAAS, particularly when exposed to changes in dietary sodium and potassium content.
Collapse
Affiliation(s)
- Anna D. Manis
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew R. Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clement J. Zablocki VA Medical Center, Milwaukee, WI 53295, USA
| |
Collapse
|
27
|
Dietary sodium modulates nephropathy in Nedd4-2-deficient mice. Cell Death Differ 2019; 27:1832-1843. [PMID: 31802037 PMCID: PMC7244563 DOI: 10.1038/s41418-019-0468-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/05/2022] Open
Abstract
Salt homeostasis is maintained by tight control of Na+ filtration and reabsorption. In the distal part of the nephron the ubiquitin protein ligase Nedd4-2 regulates membrane abundance and thus activity of the epithelial Na+ channel (ENaC), which is rate-limiting for Na+ reabsorption. Nedd4-2 deficiency in mouse results in elevated ENaC and nephropathy, however the contribution of dietary salt to this has not been characterized. In this study we show that high dietary Na+ exacerbated kidney injury in Nedd4-2-deficient mice, significantly perturbing normal postnatal nephrogenesis and resulting in multifocal areas of renal dysplasia, increased markers of kidney injury and a decline in renal function. In control mice, high dietary Na+ resulted in reduced levels of ENaC. However, Nedd4-2-deficient kidneys maintained elevated ENaC even after high dietary Na+, suggesting that the inability to efficiently downregulate ENaC is responsible for the salt-sensitivity of disease. Importantly, low dietary Na+ significantly ameliorated nephropathy in Nedd4-2-deficient mice. Our results demonstrate that due to dysregulation of ENaC, kidney injury in Nedd4-2-deficient mice is sensitive to dietary Na+, which may have implications in the management of disease in patients with kidney disease.
Collapse
|
28
|
Isaeva E, Fedoriuk M, Bohovyk R, Klemens CA, Khedr S, Golosova D, Levchenko V, El-Meanawy A, Palygin O, Staruschenko A. Vibrodissociation method for isolation of defined nephron segments from human and rodent kidneys. Am J Physiol Renal Physiol 2019; 317:F1398-F1403. [PMID: 31588797 DOI: 10.1152/ajprenal.00448.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Our current knowledge of the properties of renal ion channels responsible for electrolytes and cell energy homeostasis mainly relies on rodent studies. However, it has not been established yet to what extent their characteristics can be generalized to those of humans. The present study was designed to develop a standardized protocol for the isolation of well-preserved glomeruli and renal tubules from rodent and human kidneys and to assess the functional suitability of the obtained materials for physiological studies. Separation of nephron segments from human and rodent kidneys was achieved using a novel vibrodissociation technique. The integrity of isolated renal tubules and glomeruli was probed via electrophysiological analysis and fluorescence microscopy, and the purity of the collected fractions was confirmed using quantitative RT-PCR with gene markers for specific cell types. The developed approach allows rapid isolation of well-preserved renal tubules and glomeruli from human and rodent kidneys amenable for electrophysiological, Ca2+ imaging, and omics studies. Analysis of the basic electrophysiological parameters of major K+ and Na+ channels expressed in human cortical collecting ducts revealed that they exhibited similar biophysical properties as previously reported in rodent studies. Using vibrodissociation for nephron segment isolation has several advantages over existing techniques: it is less labor intensive, requires little to no enzymatic treatment, and produces large quantities of well-preserved experimental material in pure fractions. Applying this method for the separation of nephron segments from human and rodent kidneys may be a powerful tool for the indepth assessment of kidney function in health and disease.
Collapse
Affiliation(s)
- Elena Isaeva
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Mykhailo Fedoriuk
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Ruslan Bohovyk
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Christine A Klemens
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ashraf El-Meanawy
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
29
|
The role of distal tubule and collecting duct sodium reabsorption in sunitinib-induced hypertension. J Hypertens 2019; 36:892-903. [PMID: 29283974 DOI: 10.1097/hjh.0000000000001650] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Antiangiogenic receptor tyrosine kinase inhibitors (RTKI) induce arterial hypertension which may limit their use. Renal fractional sodium excretion (FENa) is reduced in early RTKI-induced hypertension, whereas fractional lithium excretion is unaltered. Therefore, we tested the hypothesis that activated distal tubule and collecting duct sodium reabsorption contributes to RTKI-induced hypertension. METHODS Amiloride-sensitive and hydrochlorothiazide (HCTZ)-sensitive fractional sodium reabsorption (FRNa) and renal epithelial sodium channel (ENaC) as well as sodium chloride cotransporter (NCC) abundances were determined in sunitinib-treated and control rats. The antihypertensive effects of amiloride and HCTZ were investigated by radiotelemery. RESULTS After 4 days of treatment, mean arterial pressure was 20 mmHg higher, FENa was lower (0.32 ± 0.08% vs. 0.65 ± 0.14%; P < 0.05), and renal medullary-ENaC protein abundance was higher in sunitinib-treated rats than in controls. Amiloride-sensitive FRNa was 2.37 ± 0.52% in sunitinib-treated rats vs. 2.66 ± 0.44% in controls (n.s.). HCTZ increased FENa by a similar magnitude without affecting amiloride-sensitive FRNa in both groups. After 14 days of treatment, renal medullary β-ENaC protein abundance was higher in rats that received sunitinib than in controls, whereas α-ENaC, γ-ENaC, and NCC abundances were similar in both groups. Amiloride and HCTZ reduced the sunitinib-induced mean arterial pressure rise by 8 ± 3 mmHg (P < 0.05) and 12 ± 2 mmHg (P < 0.05), respectively, without additive effects when combined. CONCLUSION ENaC-dependent and thiazide-sensitive sodium-retaining mechanisms are not overactive in sunitinib-induced hypertension but ENaC blockers and in particular thiazides may be suitable for its treatment.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The underlining goal of this review is to offer a concise, detailed look into current knowledge surrounding transient receptor potential canonical channel 6 (TRPC6) in the progression of diabetic kidney disease (DKD). RECENT FINDINGS Mutations and over-activation in TRPC6 channel activity lead to the development of glomeruli injury. Angiotensin II, reactive oxygen species, and other factors in the setting of DKD stimulate drastic increases in calcium influx through the TRPC6 channel, causing podocyte hypertrophy and foot process effacement. Loss of the podocytes further promote deterioration of the glomerular filtration barrier and play a major role in the development of both albuminuria and the renal injury in DKD. Recent genetic manipulation with TRPC6 channels in various rodent models provide additional knowledge about the role of TRPC6 in DKD and are reviewed here. The TRPC6 channel has a pronounced role in the progression of DKD, with deviations in activity yielding detrimental outcomes. The benefits of targeting TRPC6 or its upstream or downstream signaling pathways in DKD are prominent.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Clement J. Zablocki VA Medical Center, Milwaukee, WI, 53295, USA.
| | - Denisha Spires
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| |
Collapse
|
31
|
Khedr S, Palygin O, Pavlov TS, Blass G, Levchenko V, Alsheikh A, Brands MW, El-Meanawy A, Staruschenko A. Increased ENaC activity during kidney preservation in Wisconsin solution. BMC Nephrol 2019; 20:145. [PMID: 31035971 PMCID: PMC6489205 DOI: 10.1186/s12882-019-1329-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/03/2019] [Indexed: 01/09/2023] Open
Abstract
Background The invention of an effective kidney preservation solution capable of prolonging harvested kidney viability is the core of kidney transplantation procedure. Researchers have been working on upgrading the preservation solution quality aiming at prolonging storage time while maintaining utmost organ viability and functionality. For many years, the University of Wisconsin (UW) solution has been considered the gold standard solution for kidney preservation. However, the lifespan of kidney preservation in the UW solution is still limited. Its impact on the epithelial Na+ channel (ENaC) activity and its mediated processes is unknown and the primary goal of this study. Methods Kidneys harvested from 8 weeks old Sprague Dawley rats were divided into 4 groups depending upon the period of preservation in UW solution. Additional analysis was performed using dogs’ kidneys. ENaC activity was measured using patch clamp technique; protein expression and mRNA transcription were tested through Western blot and RT-qPCR, respectively. A colorimetric LDH level estimation was performed at different time points during UW solution preservation. Results Kidney preservation in Wisconsin solution caused reduction of the kidney size and weight and elevation of LDH level. ENaC activity increased in both rat and dog kidneys preserved in the UW solution as assessed by patch clamp analysis. On the contrary, ENaC channel mRNA levels remained unchanged. Conclusions ENaC activity is significantly elevated in the kidneys during preservation in UW solution, which might affect the immediate post-implantation allograft function and trajectory post-transplant.
Collapse
Affiliation(s)
- Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Present address: Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Gregory Blass
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Present address: Western Kentucky University, Bowling Green, KY, 42101, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ammar Alsheikh
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30901, USA
| | - Ashraf El-Meanawy
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Excess sodium from dietary salt (NaCl) is linked to elevations in blood pressure (BP). However, salt sensitivity of BP varies widely between individuals and there are data suggesting that salt adversely affects target organs, irrespective of BP. RECENT FINDINGS High dietary salt has been shown to adversely affect the vasculature, heart, kidneys, skin, brain, and bone. Common mediators of the target organ dysfunction include heightened inflammation and oxidative stress. These physiological alterations may contribute to disease development over time. Despite the adverse effects of salt on BP and several organ systems, there is controversy surrounding lower salt intakes and cardiovascular outcomes. Our goal here is to review the physiology contributing to BP-independent effects of salt and address the controversy around lower salt intakes and cardiovascular outcomes. We will also address the importance of background diet in modulating the effects of dietary salt.
Collapse
Affiliation(s)
- Austin T Robinson
- Department of Kinesiology and Applied Physiology, University of Delaware, 540 South College Avenue, 201M, Newark, DE, 19713, USA
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, 540 South College Avenue, 201M, Newark, DE, 19713, USA
| | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, 540 South College Avenue, 201M, Newark, DE, 19713, USA.
| |
Collapse
|
33
|
Madaio MP, Czikora I, Kvirkvelia N, McMenamin M, Yue Q, Liu T, Toque HA, Sridhar S, Covington K, Alaisami R, O'Connor PM, Caldwell RW, Chen JK, Clauss M, Brands MW, Eaton DC, Romero MJ, Lucas R. The TNF-derived TIP peptide activates the epithelial sodium channel and ameliorates experimental nephrotoxic serum nephritis. Kidney Int 2019; 95:1359-1372. [PMID: 30905471 DOI: 10.1016/j.kint.2018.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
In mice, the initial stage of nephrotoxic serum-induced nephritis (NTN) mimics antibody-mediated human glomerulonephritis. Local immune deposits generate tumor necrosis factor (TNF), which activates pro-inflammatory pathways in glomerular endothelial cells (GECs) and podocytes. Because TNF receptors mediate antibacterial defense, existing anti-TNF therapies can promote infection; however, we have previously demonstrated that different functional domains of TNF may have opposing effects. The TIP peptide mimics the lectin-like domain of TNF, and has been shown to blunt inflammation in acute lung injury without impairing TNF receptor-mediated antibacterial activity. We evaluated the impact of TIP peptide in NTN. Intraperitoneal administration of TIP peptide reduced inflammation, proteinuria, and blood urea nitrogen. The protective effect was blocked by the cyclooxygenase inhibitor indomethacin, indicating involvement of prostaglandins. Targeted glomerular delivery of TIP peptide improved pathology in moderate NTN and reduced mortality in severe NTN, indicating a local protective effect. We show that TIP peptide activates the epithelial sodium channel(ENaC), which is expressed by GEC, upon binding to the channel's α subunit. In vitro, TNF treatment of GEC activated pro-inflammatory pathways and decreased the generation of prostaglandin E2 and nitric oxide, which promote recovery from NTN. TIP peptide counteracted these effects. Despite the capacity of TIP peptide to activate ENaC, it did not increase mean arterial blood pressure in mice. In the later autologous phase of NTN, TIP peptide blunted the infiltration of Th17 cells. By countering the deleterious effects of TNF through direct actions in GEC, TIP peptide could provide a novel strategy to treat glomerular inflammation.
Collapse
Affiliation(s)
- Michael P Madaio
- Department of Medicine, Augusta University, Augusta, Georgia, USA.
| | - Istvan Czikora
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA; Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Nino Kvirkvelia
- Department of Medicine, Augusta University, Augusta, Georgia, USA
| | | | - Qiang Yue
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ting Liu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Haroldo A Toque
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | - Supriya Sridhar
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | | | - Rabei Alaisami
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Paul M O'Connor
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Robert W Caldwell
- Department of Pharmacology and Toxicology, Augusta University, Augusta, Georgia, USA
| | - Jian-Kang Chen
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Matthias Clauss
- Indiana Center for Vascular Biology and Medicine, RLR-VA Medical Center, Indianapolis, Indiana, USA
| | - Michael W Brands
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Douglas C Eaton
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maritza J Romero
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, Georgia, USA; Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Rudolf Lucas
- Department of Medicine, Augusta University, Augusta, Georgia, USA; Vascular Biology Center, Augusta University, Augusta, Georgia, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, Georgia, USA.
| |
Collapse
|
34
|
Abstract
Recent studies have suggested that postprandial increases in insulin directly contribute to reduced urinary sodium excretion. An abundance of research supports the ability of insulin to augment epithelial sodium channel (ENaC) transport. This study hypothesized that ENaC contributes to the increase in renal sodium reabsorption following a meal. To test this, we used fasted or 4 hour postprandial Sprague Dawley rats to analyze ENaC expression and activity. We also assessed total expression of additional sodium transporters (Na+-Cl− cotransporter (NCC), Na+-K+-2Cl− cotransporter (NKCC2), and Na+-K+-ATPase (NKA)) and circulating hormones involved in the renin-angiotensin-aldosterone system (RAAS). We found that after carbohydrate stimulus, ENaC open probability increased in split-open isolated collecting duct tubules, while ENaC protein levels remained unchanged. This was supported by a lack of change in phosphorylated Nedd4-2, an E3 ubiquitin ligase protein which regulates the number of ENaCs at the plasma membrane. Additionally, we found no differences in total expression of NCC, NKCC2, or NKA in the postprandial rats. Lastly, there were no significant changes in RAAS signaling between the stimulated and fasted rats, suggesting that acute hyperinsulinemia increases ENaC activity independent of the RAAS signaling cascade. These results demonstrate that insulin regulation of ENaC is a potential mechanism to preserve sodium and volume loss following a meal, and that this regulation is distinct from classical ENaC regulation by RAAS.
Collapse
|
35
|
Characterization of purinergic receptor expression in ARPKD cystic epithelia. Purinergic Signal 2018; 14:485-497. [PMID: 30417216 DOI: 10.1007/s11302-018-9632-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Polycystic kidney diseases (PKDs) are a group of inherited nephropathies marked by formation of fluid-filled cysts along the nephron. Growing evidence suggests that in the kidney formation of cysts and alteration of cystic electrolyte transport are associated with purinergic signaling. PCK/CrljCrl-Pkhd1pck/CRL (PCK) rat, an established model of autosomal recessive polycystic kidney disease (ARPKD), was used here to test this hypothesis. Cystic fluid of PCK rats and their cortical tissues exhibited significantly higher levels of ATP compared to Sprague Dawley rat kidney cortical interstitium as assessed by highly sensitive ATP enzymatic biosensors. Confocal calcium imaging of the freshly isolated cystic monolayers revealed a stronger response to ATP in a higher range of concentrations (above 100 μM). The removal of extracellular calcium results in the profound reduction of the ATP evoked transient, which suggests calcium entry into the cyst-lining cells is occurring via the extracellular (ionotropic) P2X channels. Further use of pharmacological agents (α,β-methylene-ATP, 5-BDBD, NF449, isoPPADS, AZ10606120) and immunofluorescent labeling of isolated cystic epithelia allowed us to narrow down potential candidate receptors. In conclusion, our ex vivo study provides direct evidence that the profile of P2 receptors is shifted in ARPKD cystic epithelia in an age-related manner towards prevalence of P2X4 and/or P2X7 receptors, which opens new avenues for the treatment of this disease.
Collapse
|
36
|
Ilatovskaya DV, Blass G, Palygin O, Levchenko V, Pavlov TS, Grzybowski MN, Winsor K, Shuyskiy LS, Geurts AM, Cowley AW, Birnbaumer L, Staruschenko A. A NOX4/TRPC6 Pathway in Podocyte Calcium Regulation and Renal Damage in Diabetic Kidney Disease. J Am Soc Nephrol 2018; 29:1917-1927. [PMID: 29793963 DOI: 10.1681/asn.2018030280] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022] Open
Abstract
Background Loss of glomerular podocytes is an indicator of diabetic kidney disease (DKD). The damage to these cells has been attributed in part to elevated intrarenal oxidative stress. The primary source of the renal reactive oxygen species, particularly H2O2, is NADPH oxidase 4 (NOX4). We hypothesized that NOX4-derived H2O2 contributes to podocyte damage in DKD via elevation of podocyte calcium.Methods We used Dahl salt-sensitive (SS) rats with a null mutation for the Nox4 gene (SSNox4-/-) and mice with knockout of the nonselective calcium channel TRPC6 or double knockout of TRPC5 and TRPC6. We performed whole animal studies and used biosensor measurements, electron microscopy, electrophysiology, and live calcium imaging experiments to evaluate the contribution of this pathway to the physiology of the podocytes in freshly isolated glomeruli.Results Upon induction of type 1 diabetes with streptozotocin, SSNox4-/- rats exhibited significantly lower basal intracellular Ca2+ levels in podocytes and less DKD-associated damage than SS rats did. Furthermore, the angiotensin II-elicited calcium flux was blunted in glomeruli isolated from diabetic SSNox4-/- rats compared with that in glomeruli from diabetic SS rats. H2O2 stimulated TRPC-dependent calcium influx in podocytes from wild-type mice, but this influx was blunted in podocytes from Trpc6-knockout mice and, in a similar manner, in podocytes from Trpc5/6 double-knockout mice. Finally, electron microscopy revealed that podocytes of glomeruli isolated from Trpc6-knockout or Trpc5/6 double-knockout mice were protected from damage induced by H2O2 to the same extent.Conclusions These data reveal a novel signaling mechanism involving NOX4 and TRPC6 in podocytes that could be pharmacologically targeted to abate the development of DKD.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gregory Blass
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Kristen Winsor
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Leonid S Shuyskiy
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; and.,Institute of Biomedical Research, School of Medical Sciences, Catholic University of Argentina, Buenos Aires, Argentina
| | | |
Collapse
|
37
|
Palygin O, Ilatovskaya DV, Levchenko V, Endres BT, Geurts AM, Staruschenko A. Nitric oxide production by glomerular podocytes. Nitric Oxide 2017; 72:24-31. [PMID: 29128399 DOI: 10.1016/j.niox.2017.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/27/2017] [Accepted: 11/07/2017] [Indexed: 01/18/2023]
Abstract
Nitric Oxide (NO), a potent vasodilator and vital signaling molecule, has been shown to contribute to the regulation of glomerular ultrafiltration. However, whether changes in NO occur in podocytes during the pathogenesis of salt-sensitive hypertension has not yet been thoroughly examined. We showed here that podocytes produce NO, and further hypothesized that hypertensive animals would exhibit reduced NO production in these cells in response to various paracrine factors, which might contribute to the damage of glomeruli filtration barrier and development of proteinuria. To test this, we isolated glomeruli from the kidneys of Dahl salt-sensitive (SS) rats fed a low salt (LS; 0.4% NaCl) or high salt (HS; 4% NaCl, 3 weeks) diets and loaded podocytes with either a combination of NO and Ca2+ fluorophores (DAF-FM and Fura Red, respectively) or DAF-FM alone. Changes in fluorescence were observed with confocal microscopy in response to adenosine triphosphate (ATP), angiotensin II (Ang II), and hydrogen peroxide (H2O2). Application of Ang II resulted in activation of both NO and intracellular calcium ([Ca2+]i) transients. In contrast, ATP promoted [Ca2+]i transients, but did not have any effects on NO production. SS rats fed a HS diet for 3 weeks demonstrated impaired NO production: the response to Ang II or H2O2 in podocytes of glomeruli isolated from SS rats fed a HS diet was significantly reduced compared to rats fed a LS diet. Therefore, glomerular podocytes from hypertensive rats showed a diminished NO release in response to Ang II or oxidative stress, suggesting that podocytic NO signaling is dysfunctional in this condition and likely contributes to the development of kidney injury.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bradley T Endres
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
38
|
Blass G, Levchenko V, Ilatovskaya DV, Staruschenko A. Chronic cathepsin inhibition by E-64 in Dahl salt-sensitive rats. Physiol Rep 2017; 4:4/17/e12950. [PMID: 27597769 PMCID: PMC5027357 DOI: 10.14814/phy2.12950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022] Open
Abstract
Cysteine cathepsins are lysosomal enzymes expressed in the kidneys and other tissues, and are involved in the maturation and breakdown of cellular proteins. They have been shown to be integrally involved in the progression of many cardiovascular and renal diseases. The goal of this study was to determine the involvement of cysteine cathepsins in the development of salt‐sensitive hypertension and associated kidney damage. In our experiments, Dahl salt‐sensitive (SS) rats were fed an 8% high salt NaCl diet and intravenously infused with the irreversible cysteine cathepsin inhibitor E‐64 (1 mg/day) or the vehicle (control). Both the control and E‐64 infused groups developed significant hypertension and kidney damage, and no difference of the mean arterial pressure and the hypertension‐associated albuminuria was observed between the groups. We next tested basal calcium levels in the podocytes of both control and infused groups using confocal calcium imaging. Basal calcium did not differ between the groups, indicative of the lack of a protective or aggravating influence by the cathepsin inhibition. The efficacy of E‐64 was tested in Western blotting. Our findings corresponded to the previously reported, E‐64 induced increase in cathepsin B and L abundance. We conclude that the inhibition of cysteine cathepsins by E‐64 does not have any effects on the blood pressure development and kidney damage, at least under the studied conditions of this model of SS hypertension.
Collapse
Affiliation(s)
- Gregory Blass
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
39
|
Palygin O, Levchenko V, Ilatovskaya DV, Pavlov TS, Pochynyuk OM, Jacob HJ, Geurts AM, Hodges MR, Staruschenko A. Essential role of Kir5.1 channels in renal salt handling and blood pressure control. JCI Insight 2017; 2:92331. [PMID: 28931751 PMCID: PMC5621918 DOI: 10.1172/jci.insight.92331] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 08/08/2017] [Indexed: 01/07/2023] Open
Abstract
Supplementing diets with high potassium helps reduce hypertension in humans. Inwardly rectifying K+ channels Kir4.1 (Kcnj10) and Kir5.1 (Kcnj16) are highly expressed in the basolateral membrane of distal renal tubules and contribute to Na+ reabsorption and K+ secretion through the direct control of transepithelial voltage. To define the importance of Kir5.1 in blood pressure control under conditions of salt-induced hypertension, we generated a Kcnj16 knockout in Dahl salt-sensitive (SS) rats (SSKcnj16-/-). SSKcnj16-/- rats exhibited hypokalemia and reduced blood pressure, and when fed a high-salt diet (4% NaCl), experienced 100% mortality within a few days triggered by salt wasting and severe hypokalemia. Electrophysiological recordings of basolateral K+ channels in the collecting ducts isolated from SSKcnj16-/- rats revealed activity of only homomeric Kir4.1 channels. Kir4.1 expression was upregulated in SSKcnj16-/- rats, but the protein was predominantly localized in the cytosol in SSKcnj16-/- rats. Benzamil, but not hydrochlorothiazide or furosemide, rescued this phenotype from mortality on a high-salt diet. Supplementation of high-salt diet with increased potassium (2% KCl) prevented mortality in SSKcnj16-/- rats and prevented or mitigated hypertension in SSKcnj16-/- or control SS rats, respectively. Our results demonstrate that Kir5.1 channels are key regulators of renal salt handling in SS hypertension.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology and
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | - Oleh M. Pochynyuk
- Department of Integrative Biology, University of Texas Health Science Center Medical School, Houston, Texas, USA
| | - Howard J. Jacob
- Department of Physiology and
- Human and Molecular Genetics Center and
| | - Aron M. Geurts
- Department of Physiology and
- Human and Molecular Genetics Center and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Matthew R. Hodges
- Department of Physiology and
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Alexander Staruschenko
- Department of Physiology and
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
40
|
Henshall TL, Manning JA, Alfassy OS, Goel P, Boase NA, Kawabe H, Kumar S. Deletion of Nedd4-2 results in progressive kidney disease in mice. Cell Death Differ 2017; 24:2150-2160. [PMID: 28862701 PMCID: PMC5686353 DOI: 10.1038/cdd.2017.137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/05/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
NEDD4-2 (NEDD4L), a ubiquitin protein ligase of the Nedd4 family, is a key regulator of cell surface expression and activity of the amiloride-sensitive epithelial Na+ channel (ENaC). While hypomorphic alleles of Nedd4-2 in mice show salt-sensitive hypertension, complete knockout results in pulmonary distress and perinatal lethality due to increased cell surface levels of ENaC. We now show that Nedd4-2 deficiency in mice also results in an unexpected progressive kidney injury phenotype associated with elevated ENaC and Na+Cl− cotransporter expression, increased Na+ reabsorption, hypertension and markedly reduced levels of aldosterone. The observed nephropathy is characterized by fibrosis, tubule epithelial cell apoptosis, dilated/cystic tubules, elevated expression of kidney injury markers and immune cell infiltration, characteristics reminiscent of human chronic kidney disease. Importantly, we demonstrate that the extent of kidney injury can be partially therapeutically ameliorated in mice with nephron-specific deletions of Nedd4-2 by blocking ENaC with amiloride. These results suggest that increased Na+ reabsorption via ENaC causes kidney injury and establish a novel role of NEDD4-2 in preventing Na+-induced nephropathy. Contrary to some recent reports, our data also indicate that ENaC is the primary in vivo target of NEDD4-2 and that Nedd4-2 deletion is associated with hypertension on a normal Na+ diet. These findings provide further insight into the critical function of NEDD4-2 in renal pathophysiology.
Collapse
Affiliation(s)
- Tanya L Henshall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Omri S Alfassy
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Pranay Goel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Natasha A Boase
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Hiroshi Kawabe
- Max Planck Institute of Experimental Medicine, Department of Molecular Neurobiology, Göttingen 37075, Germany
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
41
|
Wang H, Romero CA, Masjoan Juncos JX, Monu SR, Peterson EL, Carretero OA. Effect of salt intake on afferent arteriolar dilatation: role of connecting tubule glomerular feedback (CTGF). Am J Physiol Renal Physiol 2017; 313:F1209-F1215. [PMID: 28835421 DOI: 10.1152/ajprenal.00320.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/10/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023] Open
Abstract
Afferent arteriole (Af-Art) resistance is modulated by two intrinsic nephron feedbacks: 1) the vasoconstrictor tubuloglomerular feedback (TGF) mediated by Na+-K+-2Cl- cotransporters (NKCC2) in the macula densa and blocked by furosemide and 2) the vasodilator connecting tubule glomerular feedback (CTGF), mediated by epithelial Na+ channels (ENaC) in the connecting tubule and blocked by benzamil. High salt intake reduces Af-Art vasoconstrictor ability in Dahl salt-sensitive rats (Dahl SS). Previously, we measured CTGF indirectly, by differences between TGF responses with and without CTGF inhibition. We recently developed a new method to measure CTGF more directly by simultaneously inhibiting NKCC2 and the Na+/H+ exchanger (NHE). We hypothesize that in vivo during simultaneous inhibition of NKCC2 and NHE, CTGF causes an Af-Art dilatation revealed by an increase in stop-flow pressure (PSF) in Dahl SS and that is enhanced with a high salt intake. In the presence of furosemide alone, increasing nephron perfusion did not change the PSF in either Dahl salt-resistant rats (Dahl SR) or Dahl SS. When furosemide and an NHE inhibitor, dimethylamiloride, were perfused simultaneously, an increase in tubular flow caused Af-Art dilatation that was demonstrated by an increase in PSF. This increase was greater in Dahl SS [4.5 ± 0.4 (SE) mmHg] than in Dahl SR (2.5 ± 0.3 mmHg; P < 0.01). We confirmed that CTGF causes this vasodilation, since benzamil completely blocked this effect. However, a high salt intake did not augment the Af-Art dilatation. We conclude that during simultaneous inhibition of NKCC2 and NHE in the nephron, CTGF induces Af-Art dilatation and a high salt intake failed to enhance this effect.
Collapse
Affiliation(s)
- Hong Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Cesar A Romero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - J X Masjoan Juncos
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Sumit R Monu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, Michigan
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| |
Collapse
|
42
|
Wang ZR, Liu HB, Sun YY, Hu QQ, Li YX, Zheng WW, Yu CJ, Li XY, Wu MM, Song BL, Mu JJ, Yuan ZY, Zhang ZR, Ma HP. Dietary salt blunts vasodilation by stimulating epithelial sodium channels in endothelial cells from salt-sensitive Dahl rats. Br J Pharmacol 2017; 175:1305-1317. [PMID: 28409833 DOI: 10.1111/bph.13817] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/26/2017] [Accepted: 04/04/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Our recent studies show that the reduced activity of epithelial sodium channels (ENaC) in endothelial cells accounts for the adaptation of vasculature to salt in Sprague-Dawley rats. The present study examines a hypothesis that enhanced ENaC activity mediates the loss of vasorelaxation in Dahl salt-sensitive (SS) rats. EXPERIMENTAL APPROACH We used the cell-attached patch-clamp technique to record ENaC activity in split-open mesenteric arteries. Western blot and immunofluorescence staining were used to evaluate the levels of aldosterone, ENaC, eNOS and NO. Blood pressure was measured with the tail-cuff method and the artery relaxation was measured with the wire myograph assay. KEY RESULTS High-salt (HS) diet significantly increased plasma aldosterone and ENaC activity in the endothelial cells of Dahl SS rats. The endothelium-dependent artery relaxation was blunted by HS challenge in these rats. Amiloride, a potent blocker of ENaC, increased both phosphorylated eNOS and NO and therefore prevented the HS-induced loss of vasorelaxation. As, in SS rats, endogenous aldosterone was already elevated by HS challenge, exogenous aldosterone did not further elevate ENaC activity in the rats fed with HS. Eplerenone, a mineralocorticoid receptor antagonist, attenuated the effects of HS on both ENaC activity and artery relaxation. CONCLUSIONS AND IMPLICATIONS These data suggest that HS diet blunts artery relaxation and causes hypertension via a pathway associated with aldosterone-dependent activation of ENaC in endothelial cells. This pathway provides one of the mechanisms by which HS causes hypertension in Dahl SS rats. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Zi-Rui Wang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Hui-Bin Liu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacy, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying-Ying Sun
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing-Qing Hu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Yu-Xia Li
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Wei-Wan Zheng
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Chang-Jiang Yu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Xin-Yuan Li
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Ming-Ming Wu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Bin-Lin Song
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, Xi'an, China
| | - Zu-Yi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, Xi'an, China
| | - Zhi-Ren Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.,Department of Clinical Pharmacy, Institute of Clinical Pharmacy, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
43
|
Pavlov TS, Levchenko V, Ilatovskaya DV, Li H, Palygin O, Pastor-Soler NM, Hallows KR, Staruschenko A. Lack of Effects of Metformin and AICAR Chronic Infusion on the Development of Hypertension in Dahl Salt-Sensitive Rats. Front Physiol 2017; 8:227. [PMID: 28473772 PMCID: PMC5397526 DOI: 10.3389/fphys.2017.00227] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022] Open
Abstract
In the kidney, reabsorption via the epithelial sodium channel (ENaC) is involved in long-term blood pressure control. Previously we demonstrated that ENaC hyperactivity is associated with development of salt-sensitive (SS) hypertension in Dahl SS rats. AMP-activated kinase (AMPK), playing a role in cellular energy homeostasis, has been shown to decrease ENaC activity. Here, we tested whether metformin and AICAR, two drugs that activate AMPK, affect the development of salt-induced hypertension. High salt diet significantly increased mean arterial pressure (MAP) in Dahl SS rats. Blood pressure elevation was accompanied by a short-term decline of heart rate and increased circadian arterial pressure dipping. Metformin and AICAR were delivered intravenously at doses of 200 and 20 mg/kg/day, respectively. However, both control and drug-treated groups had similar development of high blood pressure within 3 weeks of 8% NaCl dietary salt intake. In the metformin-treated animals MAP reached 164.9 ± 9.1 mmHg, which was not significantly different from the control group (171.8 ± 5.6 mmHg). Patch clamp analysis revealed that the metformin-treated rats had no difference in the activity of ENaC. AICAR treatment also did not affect the development of hypertension and kidney injury. MAP reached 182.8 ± 4.8 and 178.0 ± 2.8 mmHg in AICAR and vehicle treated groups, respectively. Of note, we found that high-salt diet activated AMPK in the Dahl SS rats, and treatment with these AMPK activators had no significant further effect on AMPK activity. We conclude that AMPK activators, at least under these conditions, do not affect development of hypertension during high-salt diet in the Dahl SS rat model.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA.,Division of Hypertension and Vascular Research, Henry Ford HospitalDetroit, MI, USA
| | | | | | - Hui Li
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los AngelesLos Angeles, CA, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Nuria M Pastor-Soler
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los AngelesLos Angeles, CA, USA
| | - Kenneth R Hallows
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los AngelesLos Angeles, CA, USA
| | | |
Collapse
|
44
|
Palygin O, Pochynyuk O, Staruschenko A. Role and mechanisms of regulation of the basolateral K ir 4.1/K ir 5.1K + channels in the distal tubules. Acta Physiol (Oxf) 2017; 219:260-273. [PMID: 27129733 PMCID: PMC5086442 DOI: 10.1111/apha.12703] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/28/2016] [Accepted: 04/28/2016] [Indexed: 12/11/2022]
Abstract
Epithelial K+ channels are essential for maintaining electrolyte and fluid homeostasis in the kidney. It is recognized that basolateral inward-rectifying K+ (Kir ) channels play an important role in the control of resting membrane potential and transepithelial voltage, thereby modulating water and electrolyte transport in the distal part of nephron and collecting duct. Monomeric Kir 4.1 (encoded by Kcnj10 gene) and heteromeric Kir 4.1/Kir 5.1 (Kir 4.1 together with Kir 5.1 (Kcnj16)) channels are abundantly expressed at the basolateral membranes of the distal convoluted tubule and the cortical collecting duct cells. Loss-of-function mutations in KCNJ10 cause EAST/SeSAME tubulopathy in humans associated with salt wasting, hypomagnesaemia, metabolic alkalosis and hypokalaemia. In contrast, mice lacking Kir 5.1 have severe renal phenotype that, apart from hypokalaemia, is the opposite of the phenotype seen in EAST/SeSAME syndrome. Experimental advances using genetic animal models provided critical insights into the physiological role of these channels in electrolyte homeostasis and the control of kidney function. Here, we discuss current knowledge about K+ channels at the basolateral membrane of the distal tubules with specific focus on the homomeric Kir 4.1 and heteromeric Kir 4.1/Kir 5.1 channels. Recently identified molecular mechanisms regulating expression and activity of these channels, such as cell acidification, dopamine, insulin and insulin-like growth factor-1, Src family protein tyrosine kinases, as well as the role of these channels in NCC-mediated transport in the distal convoluted tubules, are also described.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | |
Collapse
|
45
|
Pavlov TS, Staruschenko A. Involvement of ENaC in the development of salt-sensitive hypertension. Am J Physiol Renal Physiol 2016; 313:F135-F140. [PMID: 28003189 DOI: 10.1152/ajprenal.00427.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023] Open
Abstract
Salt-sensitive hypertension is associated with renal and vascular dysfunctions, which lead to impaired fluid excretion, increased cardiac output, and total peripheral resistance. It is commonly accepted that increased renal sodium handling and plasma volume expansion are necessary factors for the development of salt-induced hypertension. The epithelial sodium channel (ENaC) is a trimeric ion channel expressed in the distal nephron that plays a critical role in the regulation of sodium reabsorption in both normal and pathological conditions. In this mini-review, we summarize recent studies investigating the role of ENaC in the development of salt-sensitive hypertension. On the basis of experimental data obtained from the Dahl salt-sensitive rats, we and others have demonstrated that abnormal ENaC activation in response to a dietary NaCl load contributes to the development of high blood pressure in this model. The role of different humoral factors, such as the components of the renin-angiotensin-aldosterone system, members of the epidermal growth factors family, arginine vasopressin, and oxidative stress mediating the effects of dietary salt on ENaC are discussed in this review to highlight future research directions and to determine potential molecular targets for drug development.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, Michigan; and
| | | |
Collapse
|
46
|
Staruschenko A, Ilatovskaya DV, Pavlov TS. High salt diet and caffeine: food for thought. J Thorac Dis 2016; 8:E1410-E1412. [PMID: 27867643 PMCID: PMC5107454 DOI: 10.21037/jtd.2016.10.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI 48202, USA
| |
Collapse
|
47
|
Ilatovskaya DV, Palygin O, Staruschenko A. Functional and therapeutic importance of purinergic signaling in polycystic kidney disease. Am J Physiol Renal Physiol 2016; 311:F1135-F1139. [PMID: 27654892 DOI: 10.1152/ajprenal.00406.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022] Open
Abstract
Polycystic kidney diseases (PKD) are a group of inherited nephropathies marked with the formation of fluid-filled cysts along the nephron. This renal disorder affects millions of people worldwide, but current treatment strategies are unfortunately limited to supportive therapy, dietary restrictions, and, eventually, renal transplantation. Recent advances in PKD management are aimed at targeting exaggerated cell proliferation and dedifferentiation to interfere with cyst growth. However, not nearly enough is known about the ion transport properties of the cystic cells, or specific signaling pathways modulating channels and transporters in this condition. There is growing evidence that abnormally elevated concentrations of adenosine triphosphate (ATP) in PKD may contribute to cyst enlargement; change in the profile of purinergic receptors may also result in promotion of cystogenesis. The current mini-review is focused on the role of ATP and associated signaling affecting ion transport properties of the renal cystic epithelia.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
48
|
Pavlov TS, Levchenko V, Ilatovskaya DV, Moreno C, Staruschenko A. Renal sodium transport in renin-deficient Dahl salt-sensitive rats. J Renin Angiotensin Aldosterone Syst 2016; 17:17/3/1470320316653858. [PMID: 27443990 PMCID: PMC5100984 DOI: 10.1177/1470320316653858] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/15/2016] [Indexed: 01/01/2023] Open
Abstract
Objective: The Dahl salt-sensitive rat is a well-established model of salt-sensitive hypertension. The goal of this study was to assess the expression and activity of renal sodium channels and transporters in the renin-deficient salt-sensitive rat. Methods: Renin knockout (Ren−/−) rats created on the salt-sensitive rat background were used to investigate the role of renin in the regulation of ion transport in salt-sensitive hypertension. Western blotting and patch-clamp analyses were utilized to assess the expression level and activity of Na+ transporters. Results: It has been described previously that Ren−/− rats exhibit severe kidney underdevelopment, polyuria, and lower body weight and blood pressure compared to their wild-type littermates. Here we found that renin deficiency led to decreased expression of sodium-hydrogen antiporter (NHE3), the Na+/H+ exchanger involved in Na+ absorption in the proximal tubules, but did not affect the expression of Na-K-Cl cotransporter (NKCC2), the main transporter in the loop of Henle. In the distal nephron, the expression of sodium chloride cotransporter (NCC) was lower in Ren−/− rats. Single-channel patch clamp analysis detected decreased ENaC activity in Ren−/− rats which was mediated via changes in the channel open probability. Conclusion: These data illustrate that renin deficiency leads to significant dysregulation of ion transporters.
Collapse
Affiliation(s)
| | | | | | - Carol Moreno
- Department of Physiology, Medical College of Wisconsin, USA Cardiovascular and Metabolic Diseases, MedImmune, Cambridge, UK
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, USA Cardiovascular Center, Medical College of Wisconsin, USA
| |
Collapse
|
49
|
Affiliation(s)
- John E Hall
- From the Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson.
| |
Collapse
|
50
|
Chen J, Zeng F, Forrester SJ, Eguchi S, Zhang MZ, Harris RC. Expression and Function of the Epidermal Growth Factor Receptor in Physiology and Disease. Physiol Rev 2016; 96:1025-1069. [DOI: 10.1152/physrev.00030.2015] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is the prototypical member of a family of membrane-associated intrinsic tyrosine kinase receptors, the ErbB family. EGFR is activated by multiple ligands, including EGF, transforming growth factor (TGF)-α, HB-EGF, betacellulin, amphiregulin, epiregulin, and epigen. EGFR is expressed in multiple organs and plays important roles in proliferation, survival, and differentiation in both development and normal physiology, as well as in pathophysiological conditions. In addition, EGFR transactivation underlies some important biologic consequences in response to many G protein-coupled receptor (GPCR) agonists. Aberrant EGFR activation is a significant factor in development and progression of multiple cancers, which has led to development of mechanism-based therapies with specific receptor antibodies and tyrosine kinase inhibitors. This review highlights the current knowledge about mechanisms and roles of EGFR in physiology and disease.
Collapse
Affiliation(s)
- Jianchun Chen
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Fenghua Zeng
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Steven J. Forrester
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ming-Zhi Zhang
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Raymond C. Harris
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|