1
|
Soleman SI, Maya J, Levesque P, Mohammad A, Christopher L, Schumacher J, Nanduri A, Sivakumar P, Kozinn M, Costet P, Wang C, Richter J, Hawthorne D, Bui A, Rao VS, Dickerson D, Testani J, Ramírez-Valle F, Baribaud F, Murthy B, Merali S. First-in-Human Study to Assess the Safety, Pharmacokinetics, and Pharmacodynamics of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor. Clin Pharmacol Ther 2024; 116:1627-1634. [PMID: 39219444 DOI: 10.1002/cpt.3430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
In patients with heart failure (HF) who respond inadequately to loop diuretic therapy, BMS-986308, an oral, selective, reversible renal outer medullary potassium channel (ROMK) inhibitor may represent an effective diuretic with a novel mechanism of action. We present data from the first-in-human study aimed to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) following single ascending doses of BMS-986308 in healthy adult participants. Forty healthy participants, aged from 20 to 55 years, and body mass index (BMI) from 19.8 to 31.6 kg/m2 were assigned to 1 of 5 dose cohorts (1, 3, 10, 30, and 100 mg) and randomized (6:2) to receive BMS-986308 oral solution or matching placebo. Following administration, BMS-986308 was rapidly absorbed with a median time to maximum concentration (Tmax) of 1.00 to 1.75 h and exhibiting a mean terminal half-life (t1/2) of approximately 13 h. Dose proportionality was evident in BMS-986308 area under the concentration-time curve (AUC), while maximum concentration (Cmax) was slightly greater than dose-proportional. We observed that urine output (or diuresis; mL) and urinary sodium excretion (or natriuresis; mmol) increased in a dose-dependent manner, starting at a minimum pharmacologically active dose of 30 mg. The largest mean changes from baseline in diuresis and natriuresis occurred in both the 6- and -24 h post-dose period following administration of 100 mg (1683.0 mL and 2055.3 mL, and 231.7 mmol and 213.7 mmol, respectively; ***P < 0.001). Overall, single-dose BMS-986308 was found to be safe, well-tolerated, with an excellent PK profile, and substantial diuretic and natriuretic activity.
Collapse
Affiliation(s)
- Sharif I Soleman
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Juan Maya
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Paul Levesque
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Atif Mohammad
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Lisa Christopher
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Justin Schumacher
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Aparna Nanduri
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | | | - Marc Kozinn
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Philippe Costet
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Chang Wang
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Jeremy Richter
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Dara Hawthorne
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Anh Bui
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Veena S Rao
- Department of Cardiovascular Medicine, Yale Medical Center, New Haven, Connecticut, USA
| | | | - Jeffrey Testani
- Department of Cardiovascular Medicine, Yale Medical Center, New Haven, Connecticut, USA
| | | | - Frédéric Baribaud
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Bindu Murthy
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| | - Samira Merali
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey, USA
| |
Collapse
|
2
|
Schary N, Edemir B, Todorov VT. A Possible Link between Cell Plasticity and Renin Expression in the Collecting Duct: A Narrative Review. Int J Mol Sci 2024; 25:9549. [PMID: 39273497 PMCID: PMC11395489 DOI: 10.3390/ijms25179549] [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: 08/01/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
The hormone renin is produced in the kidney by the juxtaglomerular cells. It is the rate-limiting factor in the circulating renin-angiotensin-aldosterone system (RAAS), which contributes to electrolyte, water, and blood pressure homeostasis. In the kidneys, the distal tubule and the collecting duct are the key target segments for RAAS. The collecting duct is important for urine production and also for salt, water, and acid-base homeostasis. The critical functional role of the collecting duct is mediated by the principal and the intercalated cells and is regulated by different hormones like aldosterone and vasopressin. The collecting duct is not only a target for hormones but also a place of hormone production. It is accepted that renin is produced in the collecting duct at a low level. Several studies have described that the cells in the collecting duct exhibit plasticity properties because the ratio of principal to intercalated cells can change under specific circumstances. This narrative review focuses on two aspects of the collecting duct that remain somehow aside from mainstream research, namely the cell plasticity and the renin expression. We discuss the link between these collecting duct features, which we see as a promising area for future research given recent findings.
Collapse
Affiliation(s)
- Nicole Schary
- Department of Physiology and Pathophysiology, Center of Biomedical Education and Research (ZBAF), Faculty of Health—School of Medicine, Witten/Herdecke University, 58453 Witten, Germany;
| | - Bayram Edemir
- Department of Physiology and Pathophysiology, Center of Biomedical Education and Research (ZBAF), Faculty of Health—School of Medicine, Witten/Herdecke University, 58453 Witten, Germany;
- Department of Internal Medicine IV, Hematology and Oncology, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Vladimir T. Todorov
- Department of Physiology and Pathophysiology, Center of Biomedical Education and Research (ZBAF), Faculty of Health—School of Medicine, Witten/Herdecke University, 58453 Witten, Germany;
- Experimental Nephrology and Division of Nephrology, Department of Internal Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
3
|
Duan XP, Zhang CB, Wang WH, Lin DH. Role of calcineurin in regulating renal potassium (K +) excretion: Mechanisms of calcineurin inhibitor-induced hyperkalemia. Acta Physiol (Oxf) 2024; 240:e14189. [PMID: 38860527 PMCID: PMC11250626 DOI: 10.1111/apha.14189] [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: 03/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Calcineurin, protein phosphatase 2B (PP2B) or protein phosphatase 3 (PP3), is a calcium-dependent serine/threonine protein phosphatase. Calcineurin is widely expressed in the kidney and regulates renal Na+ and K+ transport. In the thick ascending limb, calcineurin plays a role in inhibiting NKCC2 function by promoting the dephosphorylation of the cotransporter and an intracellular sorting receptor, called sorting-related-receptor-with-A-type repeats (SORLA), is involved in modulating the effect of calcineurin on NKCC2. Calcineurin also participates in regulating thiazide-sensitive NaCl-cotransporter (NCC) in the distal convoluted tubule. The mechanisms by which calcineurin regulates NCC include directly dephosphorylation of NCC, regulating Kelch-like-3/CUL3 E3 ubiquitin-ligase complex, which is responsible for WNK (with-no-lysin-kinases) ubiquitination, and inhibiting Kir4.1/Kir5.1, which determines NCC expression/activity. Finally, calcineurin is also involved in regulating ROMK (Kir1.1) channels in the cortical collecting duct and Cyp11 2 expression in adrenal zona glomerulosa. In summary, calcineurin is involved in the regulation of NKCC2, NCC, and inwardly rectifying K+ channels in the kidney, and it also plays a role in modulating aldosterone synthesis in adrenal gland, which regulates epithelial-Na+-channel expression/activity. Thus, application of calcineurin inhibitors (CNIs) is expected to abrupt calcineurin-mediated regulation of transepithelial Na+ and K+ transport in the kidney. Consequently, CNIs cause hypertension, compromise renal K+ excretion, and induce hyperkalemia.
Collapse
Affiliation(s)
- Xin-Peng Duan
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Biao Zhang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
4
|
Wu P, Li ST, Shu TT, Mao ZH, Fu WJ, Yang YY, Pan SK, Liu DW, Liu ZS, Gao ZX. Impaired distal renal potassium handling in streptozotocin-induced diabetic mice. Am J Physiol Renal Physiol 2024; 327:F158-F170. [PMID: 38779755 DOI: 10.1152/ajprenal.00240.2023] [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: 08/14/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Diabetes is closely associated with K+ disturbances during disease progression and treatment. However, it remains unclear whether K+ imbalance occurs in diabetes with normal kidney function. In this study, we examined the effects of dietary K+ intake on systemic K+ balance and renal K+ handling in streptozotocin (STZ)-induced diabetic mice. The control and STZ mice were fed low or high K+ diet for 7 days to investigate the role of dietary K+ intake in renal K+ excretion and K+ homeostasis and to explore the underlying mechanism by evaluating K+ secretion-related transport proteins in distal nephrons. K+-deficient diet caused excessive urinary K+ loss, decreased daily K+ balance, and led to severe hypokalemia in STZ mice compared with control mice. In contrast, STZ mice showed an increased daily K+ balance and elevated plasma K+ level under K+-loading conditions. Dysregulation of the NaCl cotransporter (NCC), epithelial Na+ channel (ENaC), and renal outer medullary K+ channel (ROMK) was observed in diabetic mice fed either low or high K+ diet. Moreover, amiloride treatment reduced urinary K+ excretion and corrected hypokalemia in K+-restricted STZ mice. On the other hand, inhibition of SGLT2 by dapagliflozin promoted urinary K+ excretion and normalized plasma K+ levels in K+-supplemented STZ mice, at least partly by increasing ENaC activity. We conclude that STZ mice exhibited abnormal K+ balance and impaired renal K+ handling under either low or high K+ diet, which could be primarily attributed to the dysfunction of ENaC-dependent renal K+ excretion pathway, despite the possible role of NCC.NEW & NOTEWORTHY Neither low dietary K+ intake nor high dietary K+ intake effectively modulates renal K+ excretion and K+ homeostasis in STZ mice, which is closely related to the abnormality of ENaC expression and activity. SGLT2 inhibitor increases urinary K+ excretion and reduces plasma K+ level in STZ mice under high dietary K+ intake, an effect that may be partly due to the upregulation of ENaC activity.
Collapse
Affiliation(s)
- Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Shu-Ting Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Ting-Ting Shu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Wen-Jia Fu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Yuan-Yuan Yang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| | - Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, People's Republic of China
| |
Collapse
|
5
|
Richter JM, Gunaga P, Yadav N, Bora RO, Bhide R, Rajugowda N, Govindrajulu K, Godesi S, Akuthota N, Rao P, Sivaraman A, Panda M, Kaspady M, Gupta A, Mathur A, Levesque PC, Gulia J, Dokania M, Ramarao M, Kole P, Chacko S, Lentz KA, Sivaprasad Lvj S, Thatipamula RP, Sridhar S, Kamble S, Govindrajan A, Soleman SI, Gordon DA, Wexler RR, Priestley ES. Discovery of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor for the Treatment of Heart Failure. J Med Chem 2024; 67:9731-9744. [PMID: 38807539 DOI: 10.1021/acs.jmedchem.4c00893] [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: 05/30/2024]
Abstract
Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.
Collapse
Affiliation(s)
- Jeremy M Richter
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashantha Gunaga
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Navnath Yadav
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajesh Onkardas Bora
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajeev Bhide
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Nagendra Rajugowda
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Kavitha Govindrajulu
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sreenivasulu Godesi
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Nagarjuna Akuthota
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Prasanna Rao
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Aneesh Sivaraman
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoranjan Panda
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Mahammed Kaspady
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Anuradha Gupta
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Paul C Levesque
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Jyoti Gulia
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoj Dokania
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manjunath Ramarao
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashant Kole
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Silvi Chacko
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Kimberley A Lentz
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Sankara Sivaprasad Lvj
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | | | - Srikanth Sridhar
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Shyam Kamble
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Arun Govindrajan
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sharif I Soleman
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - David A Gordon
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Ruth R Wexler
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - E Scott Priestley
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| |
Collapse
|
6
|
Guo L, Han J, Wang Y, Chang Y, Qu W, Man C, Fei P, Jiang Y. Antibacterial action of slightly acidic electrolytic water against Cronobacter sakazakii and its application as a disinfectant on high-risk contact surfaces. Front Microbiol 2024; 15:1314362. [PMID: 38351917 PMCID: PMC10864107 DOI: 10.3389/fmicb.2024.1314362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Powdered infant formula (PIF) is prone to Cronobacter sakazakii (C. sakazakii) contamination, which can result in infections that endanger the lives of newborns and infants. Slightly acidic electrolytic water (SAEW) has shown antibacterial effects on a variety of foodborne pathogens and has a wide applicability in the food industry. Here, the antibacterial activity of SAEW against C. sakazakii and its use as a disinfectant on contact surfaces with high infection transmission risk were investigated. The inactivation of SAEW on C. sakazakii was positively correlated to the SAEW concentration and treatment time. The antibacterial effect of SAEW was achieved by decreasing the intracellular adenosine triphosphate (ATP), K+, protein, and DNA contents of C. sakazakii, reducing the intracellular pH (pHin) and destroying the cell morphology, which led to inactivation of C. sakazakii ultimately. To test the applicability of this study, the results showed that approximately 103 CFU/cm2 of C. sakazakii were successfully inactivated on stainless steel and rubber surfaces after a 30 mg/L SAEW treatment for 20 s. These results indicate the antibacterial mechanism and potential application of SAEW against C. sakazakii, as well as a new strategy for the prevention and control of C. sakazakii on stainless steel and rubber surfaces.
Collapse
Affiliation(s)
- Ling Guo
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
- Food Laboratory of Zhongyuan, Luohe, China
| | - Jing Han
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Yanyan Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Yajing Chang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Wenxuan Qu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Peng Fei
- School of Zhang Zhongjing Health Care and Food, Nanyang Institute of Technology, Nanyang, China
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
7
|
Duan XP, Xiao Y, Su XT, Zheng JY, Gurley S, Emathinger J, Yang CL, McCormick J, Ellison DH, Lin DH, Wang WH. Role of Angiotensin II Type 1a Receptor (AT1aR) of Renal Tubules in Regulating Inwardly Rectifying Potassium Channels 4.2 (Kir4.2), Kir4.1, and Epithelial Na + Channel (ENaC). Hypertension 2024; 81:126-137. [PMID: 37909221 PMCID: PMC10842168 DOI: 10.1161/hypertensionaha.123.21389] [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: 04/19/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Kir4.2 and Kir4.1 play a role in regulating membrane transport in the proximal tubule (PT) and in the distal-convoluted-tubule (DCT), respectively. METHODS We generated kidney-tubule-specific-AT1aR-knockout (Ks-AT1aR-KO) mice to examine whether renal AT1aR regulates Kir4.2 and Kir4.1. RESULTS Ks-AT1aR-KO mice had a lower systolic blood pressure than Agtr1aflox/flox (control) mice. Ks-AT1aR-KO mice had a lower expression of NHE3 (Na+/H+-exchanger 3) and Kir4.2, a major Kir-channel in PT, than Agtr1aflox/flox mice. Whole-cell recording also demonstrated that the membrane potential in PT of Ks-AT1aR-KO mice was lesser negative than Agtr1aflox/flox mice. The expression of Kir4.1 and Kir5.1, Kir4.1/Kir5.1-mediated K+ currents of DCT and DCT membrane potential in Ks-AT1aR-KO mice, were similar to Agtr1aflox/flox mice. However, angiotensin II perfusion for 7 days hyperpolarized the membrane potential in PT and DCT of the control mice but not in Ks-AT1aR-KO mice, while angiotensin II perfusion did not change the expression of Kir4.1, Kir4.2, and Kir5.1. Deletion of AT1aR did not significantly affect the expression of αENaC (epithelial Na+ channel) and βENaC but increased cleaved γENaC expression. Patch-clamp experiments demonstrated that deletion of AT1aR increased amiloride-sensitive Na+-currents in the cortical-collecting duct but not in late-DCT. However, tertiapin-Q sensitive renal outer medullary potassium channel currents were similar in both genotypes. CONCLUSIONS AT1aR determines the baseline membrane potential of PT by controlling Kir4.2 expression/activity but AT1aR is not required for determining the baseline membrane potential of the DCT and Kir4.1/Kir5.1 activity/expression. However, AT1aR is required for angiotensin II-induced hyperpolarization of basolateral membrane of PT and DCT. Deletion of AT1aR had no effect on baseline renal outer medullary potassium channel activity but increased ENaC activity in the CCD.
Collapse
Affiliation(s)
- Xin-Peng Duan
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Yu Xiao
- Department of Physiology, Qiqihar Medical College, Heilongjiang, China
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Xiao-Tong Su
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Jun-Ya Zheng
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Susan Gurley
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | | | - Chao-Ling Yang
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - James McCormick
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - David H. Ellison
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY
| |
Collapse
|
8
|
Li C, Yang Y. Advancements in the study of inward rectifying potassium channels on vascular cells. Channels (Austin) 2023; 17:2237303. [PMID: 37463317 PMCID: PMC10355679 DOI: 10.1080/19336950.2023.2237303] [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: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/04/2023] [Indexed: 07/20/2023] Open
Abstract
Inward rectifier potassium channels (Kir channels) exist in a variety of cells and are involved in maintaining resting membrane potential and signal transduction in most cells, as well as connecting metabolism and membrane excitability of body cells. It is closely related to normal physiological functions of body and the occurrence and development of some diseases. Although the functional expression of Kir channels and their role in disease have been studied, they have not been fully elucidated. In this paper, the functional expression of Kir channels in vascular endothelial cells and smooth muscle cells and their changes in disease states were reviewed, especially the recent research progress of Kir channels in stem cells was introduced, in order to have a deeper understanding of Kir channels in vascular tissues and provide new ideas and directions for the treatment of related ion channel diseases.
Collapse
Affiliation(s)
- Chunshu Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
9
|
Orfali R, AlFaiz A, Rahman MA, Lau L, Nam YW, Zhang M. K Ca2 and K Ca3.1 Channels in the Airways: A New Therapeutic Target. Biomedicines 2023; 11:1780. [PMID: 37509419 PMCID: PMC10376499 DOI: 10.3390/biomedicines11071780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
K+ channels are involved in many critical functions in lung physiology. Recently, the family of Ca2+-activated K+ channels (KCa) has received more attention, and a massive amount of effort has been devoted to developing selective medications targeting these channels. Within the family of KCa channels, three small-conductance Ca2+-activated K+ (KCa2) channel subtypes, together with the intermediate-conductance KCa3.1 channel, are voltage-independent K+ channels, and they mediate Ca2+-induced membrane hyperpolarization. Many KCa2 channel members are involved in crucial roles in physiological and pathological systems throughout the body. In this article, different subtypes of KCa2 and KCa3.1 channels and their functions in respiratory diseases are discussed. Additionally, the pharmacology of the KCa2 and KCa3.1 channels and the link between these channels and respiratory ciliary regulations will be explained in more detail. In the future, specific modulators for small or intermediate Ca2+-activated K+ channels may offer a unique therapeutic opportunity to treat muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Ali AlFaiz
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Liz Lau
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
10
|
Janjic P, Solev D, Kocarev L. Non-trivial dynamics in a model of glial membrane voltage driven by open potassium pores. Biophys J 2023; 122:1470-1490. [PMID: 36919241 PMCID: PMC10147837 DOI: 10.1016/j.bpj.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/01/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Despite the molecular evidence that a nearly linear steady-state current-voltage relationship in mammalian astrocytes reflects a total current resulting from more than one differentially regulated K+ conductance, detailed ordinary differential equation (ODE) models of membrane voltage Vm are still lacking. Various experimental results reporting altered rectification of the major Kir currents in glia, dominated by Kir4.1, have motivated us to develop a detailed model of Vm dynamics incorporating the weaker potassium K2P-TREK1 current in addition to Kir4.1, and study the stability of the resting state Vr. The main question is whether, with the loss of monotonicity in glial I-V curve resulting from altered Kir rectification, the nominal resting state Vr remains stable, and the cell retains the trivial, potassium electrode behavior with Vm after EK. The minimal two-dimensional model of Vm near Vr showed that an N-shape deformed Kir I-V curve induces multistability of Vm in a model that incorporates K2P activation kinetics, and nonspecific K+ leak currents. More specifically, an asymmetrical, nonlinear decrease of outward Kir4.1 conductance, turning the channels into inward rectifiers, introduces instability of Vr. That happens through a robust bifurcation giving birth to a second, more depolarized stable resting state Vdr > -10 mV. Realistic recordings from electrographic seizures were used to perturb the model. Simulations of the model perturbed by constant current through gap junctions and seizure-like discharges as local field potentials led to depolarization and switching of Vm between the two stable states, in a downstate-upstate manner. In the event of prolonged depolarizations near Vdr, such catastrophic instability would affect all aspects of the glial function, from metabolic support to membrane transport, and practically all neuromodulatory roles assigned to glia.
Collapse
Affiliation(s)
- Predrag Janjic
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia.
| | - Dimitar Solev
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| | - Ljupco Kocarev
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| |
Collapse
|
11
|
Vieira-Coelho MA, Martel F. Inhibition of kidney potassium channels by fluoxetine: In vivo and in vitro studies. Fundam Clin Pharmacol 2023; 37:226-234. [PMID: 36103995 DOI: 10.1111/fcp.12833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
In vitro studies have demonstrated that fluoxetine, a commonly used antidepressant drug, can modulate the activity of K+ channels. In the present study, we investigated the in vivo effect of acute and sub-chronic treatment of rats with fluoxetine on K+ renal transport. Furthermore, OK cells, a kidney epithelial cell line, were used in order to evaluate the in vitro effect of fluoxetine on K+ currents. In the sub-chronic study, fluoxetine was administrated daily (10 mg/kg, p.o.) for 15 days to male adult Wistar rats. In the acute study, rats were given increasing doses of fluoxetine (1, 3, 10, 30 and 50 mg/kg, p.o.) for 24 h. Results from the sub-chronic study show that urinary K+ content (in mmol/L) was markedly reduced in the fluoxetine-treated animals (fluoxetine: 83 ± 9; control: 131 ± 10; P < 0.001). K+ fractional renal excretion (in %) was also significantly lower in the fluoxetine group (fluoxetine: 6 ± 1; control: 13 ± 2; P < 0.001). No significant changes was observed in creatinine clearance and on renal tubular Na+ ,K+ -ATPase activity. Results obtained from the acute study demonstrate that, after a 24-h administration, fluoxetine produced a dose-dependent decrease in urinary K+ , with an ED50 (in mg/kg) of 4.2 (2.8; 5.5) and a maximal effect of 62% reduction. In vitro, fluoxetine produced a concentration-dependent inhibition of K+ currents in OK cells, with an EC50 of 107 (84.8; 129.5) μM. In conclusion, fluoxetine produces a marked reduction on urinary K+ excretion; this effect constitutes an in vivo evidence for the inhibitory action of fluoxetine on kidney epithelial K+ channels.
Collapse
Affiliation(s)
- Maria A Vieira-Coelho
- Unit of Pharmacology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, University of Porto, Porto, Portugal
| |
Collapse
|
12
|
Johnston JG, Welch AK, Cain BD, Sayeski PP, Gumz ML, Wingo CS. Aldosterone: Renal Action and Physiological Effects. Compr Physiol 2023; 13:4409-4491. [PMID: 36994769 PMCID: PMC11472823 DOI: 10.1002/cphy.c190043] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Aldosterone exerts profound effects on renal and cardiovascular physiology. In the kidney, aldosterone acts to preserve electrolyte and acid-base balance in response to changes in dietary sodium (Na+ ) or potassium (K+ ) intake. These physiological actions, principally through activation of mineralocorticoid receptors (MRs), have important effects particularly in patients with renal and cardiovascular disease as demonstrated by multiple clinical trials. Multiple factors, be they genetic, humoral, dietary, or otherwise, can play a role in influencing the rate of aldosterone synthesis and secretion from the adrenal cortex. Normally, aldosterone secretion and action respond to dietary Na+ intake. In the kidney, the distal nephron and collecting duct are the main targets of aldosterone and MR action, which stimulates Na+ absorption in part via the epithelial Na+ channel (ENaC), the principal channel responsible for the fine-tuning of Na+ balance. Our understanding of the regulatory factors that allow aldosterone, via multiple signaling pathways, to function properly clearly implicates this hormone as central to many pathophysiological effects that become dysfunctional in disease states. Numerous pathologies that affect blood pressure (BP), electrolyte balance, and overall cardiovascular health are due to abnormal secretion of aldosterone, mutations in MR, ENaC, or effectors and modulators of their action. Study of the mechanisms of these pathologies has allowed researchers and clinicians to create novel dietary and pharmacological targets to improve human health. This article covers the regulation of aldosterone synthesis and secretion, receptors, effector molecules, and signaling pathways that modulate its action in the kidney. We also consider the role of aldosterone in disease and the benefit of mineralocorticoid antagonists. © 2023 American Physiological Society. Compr Physiol 13:4409-4491, 2023.
Collapse
Affiliation(s)
- Jermaine G Johnston
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Amanda K Welch
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Peter P Sayeski
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| |
Collapse
|
13
|
Meng XX, Zhang H, Meng GL, Jiang SP, Duan XP, Wang WH, Wang MX. The effect of high-dietary K + (HK) on Kir4.1/Kir5.1 and ROMK in the distal convoluted tubule (DCT) is not affected by gender and Cl - content of the diet. Front Physiol 2022; 13:1039029. [PMID: 36439248 PMCID: PMC9682262 DOI: 10.3389/fphys.2022.1039029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Basolateral potassium channels in the distal convoluted tubule (DCT) are composed of inwardly-rectifying potassium channel 4.1 (Kir4.1) and Kir5.1. Kir4.1 interacts with Kir5.1 to form a 40 pS K+ channel which is the only type K+ channel expressed in the basolateral membrane of the DCT. Moreover, Kir4.1/Kir5.1 heterotetramer plays a key role in determining the expression and activity of thiazide-sensitive Na-Cl cotransport (NCC). In addition to Kir4.1/Kir5.1, Kir1.1 (ROMK) is expressed in the apical membrane of the late DCT (DCT2) and plays a key role in mediating epithelial Na+ channel (ENaC)-dependent K+ excretion. High dietary-K+-intake (HK) stimulates ROMK and inhibits Kir4.1/Kir5.1 in the DCT. Inhibition of Kir4.1/Kir5.1 is essential for HK-induced suppression of NCC whereas the stimulation of ROMK is important for increasing ENaC-dependent K+ excretion during HK. We have now used the patch-clamp-technique to examine whether gender and Cl- content of K+-diet affect HK-induced inhibition of basolateral Kir4.1/Kir5.1 and HK-induced stimulation of ROMK. Single-channel-recording shows that basolateral 40 pS K+ channel (Kir4.1/Kir5.1) activity of the DCT defined by NPo was 1.34 (1% KCl, normal K, NK), 0.95 (5% KCl) and 1.03 (5% K+-citrate) in male mice while it was 1.47, 1.02 and 1.05 in female mice. The whole-cell recording shows that Kir4.1/Kir5.1-mediated-K+ current of the early-DCT (DCT1) was 1,170 pA (NK), 725 pA (5% KCl) and 700 pA (5% K+-citrate) in male mice whereas it was 1,125 pA, 674 pA and 700 pA in female mice. Moreover, K+-currents (IK) reversal potential of DCT (an index of membrane potential) was -63 mV (NK), -49 mV (5% KCl) and -49 mV (5% K-citrate) in the male mice whereas it was -63 mV, -50 mV and -50 mV in female mice. Finally, TPNQ-sensitive whole-cell ROMK-currents in the DCT2 /initial-connecting tubule (CNT) were 910 pA (NK), 1,520 pA (5% KCl) and 1,540 pA (5% K+-citrate) in male mice whereas the ROMK-mediated K+ currents were 1,005 pA, 1,590 pA and 1,570 pA in female mice. We conclude that the effect of HK intake on Kir4.1/Kir5.1 of the DCT and ROMK of DCT2/CNT is similar between male and female mice. Also, Cl- content in HK diets has no effect on HK-induced inhibition of Kir4.1/Kir5.1 of the DCT and HK-induced stimulation of ROMK in DCT2/CNT.
Collapse
Affiliation(s)
- Xin-Xin Meng
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Hao Zhang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Gui-Lin Meng
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Shao-Peng Jiang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xin-Peng Duan
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States,*Correspondence: Ming-Xiao Wang, ; Wen-Hui Wang,
| | - Ming-Xiao Wang
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China,*Correspondence: Ming-Xiao Wang, ; Wen-Hui Wang,
| |
Collapse
|
14
|
Lo J, Forst AL, Warth R, Zdebik AA. EAST/SeSAME Syndrome and Beyond: The Spectrum of Kir4.1- and Kir5.1-Associated Channelopathies. Front Physiol 2022; 13:852674. [PMID: 35370765 PMCID: PMC8965613 DOI: 10.3389/fphys.2022.852674] [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: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
In 2009, two groups independently linked human mutations in the inwardly rectifying K+ channel Kir4.1 (gene name KCNJ10) to a syndrome affecting the central nervous system (CNS), hearing, and renal tubular salt reabsorption. The autosomal recessive syndrome has been named EAST (epilepsy, ataxia, sensorineural deafness, and renal tubulopathy) or SeSAME syndrome (seizures, sensorineural deafness, ataxia, intellectual disability, and electrolyte imbalance), accordingly. Renal dysfunction in EAST/SeSAME patients results in loss of Na+, K+, and Mg2+ with urine, activation of the renin-angiotensin-aldosterone system, and hypokalemic metabolic alkalosis. Kir4.1 is highly expressed in affected organs: the CNS, inner ear, and kidney. In the kidney, it mostly forms heteromeric channels with Kir5.1 (KCNJ16). Biallelic loss-of-function mutations of Kir5.1 can also have disease significance, but the clinical symptoms differ substantially from those of EAST/SeSAME syndrome: although sensorineural hearing loss and hypokalemia are replicated, there is no alkalosis, but rather acidosis of variable severity; in contrast to EAST/SeSAME syndrome, the CNS is unaffected. This review provides a framework for understanding some of these differences and will guide the reader through the growing literature on Kir4.1 and Kir5.1, discussing the complex disease mechanisms and the variable expression of disease symptoms from a molecular and systems physiology perspective. Knowledge of the pathophysiology of these diseases and their multifaceted clinical spectrum is an important prerequisite for making the correct diagnosis and forms the basis for personalized therapies.
Collapse
Affiliation(s)
- Jacky Lo
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Anna-Lena Forst
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Richard Warth
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Anselm A. Zdebik
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- Centre for Nephrology, University College London, London, United Kingdom
| |
Collapse
|
15
|
Evolving concepts of TRPV4 in controlling flow-sensitivity of the renal nephron. CURRENT TOPICS IN MEMBRANES 2022; 89:75-94. [DOI: 10.1016/bs.ctm.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Unappreciated Roles for K+ Channels in Bacterial Physiology. Trends Microbiol 2021; 29:942-950. [DOI: 10.1016/j.tim.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 01/03/2023]
|
17
|
Zhang G, Lv Z, Zhao Y, Chen R, Zhan X, Wang W, Sui H. Inhibitory effect of tumor necrosis factor-α on the basolateral Kir4.1/Kir5.1 channels in the thick ascending limb during diabetes. Exp Ther Med 2021; 22:1242. [PMID: 34539838 DOI: 10.3892/etm.2021.10677] [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: 04/30/2021] [Accepted: 08/13/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetic nephropathy is a major contributor to the morbidity and mortality of patients with diabetes. TNF-α expression is elevated during diabetes and is implicated in the pathogenesis of diabetic nephropathy; however, its underlying molecular mechanisms remain unclear. The present study aimed to investigate the effect and molecular mechanism of TNF-α on the basolateral inwardly rectifying potassium (Kir)4.1/Kir5.1 channels in the thick ascending limb (TAL) of rat kidneys using western blotting and the patch clamp technique to provide a theoretical basis for the cause of the decrease in kidney concentrating capacity during diabetes. The results demonstrated that urinary TNF-α excretion and protein TNF-α expression in the TAL increased and basolateral Kir4.1/Kir5.1 channel activity decreased during diabetes; however, diabetic rats exhibited amelioration of Kir4.1/Kir5.1 activity with a soluble TNF-α antagonist, TNF receptor fusion protein (TNFR:Fc). These results suggested that TNF-α inhibited the activity of the basolateral Kir4.1/Kir5.1 channel in the TAL of rat kidneys during diabetes. In addition, the protein expression levels of phospholipase A2 (PLA2) and cyclooxygenase-2 (COX2) increased in diabetic rats, the effects of which deceased following treatment with TNFR:Fc compared with the diabetic group. Furthermore, an agonist of PLA2 (melittin) and COX2 production [prostaglandin E2 (PGE2)] inhibited the basolateral Kir4.1/Kir5.1 channels. Taken together, the results of the present study suggested that the inhibitory effect of TNF-α on the basolateral Kir4.1/Kir5.1 channels in the TAL during diabetes is mediated by the PLA2/COX2/PGE2 signaling pathway.
Collapse
Affiliation(s)
- Guoyan Zhang
- Department of Urology and Endocrinology, First Affiliated Hospital, Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Zhiming Lv
- Department of Urology and Endocrinology, First Affiliated Hospital, Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Yang Zhao
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Rui Chen
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Xiangyu Zhan
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Weiqun Wang
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Hongyu Sui
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| |
Collapse
|
18
|
Kir Channel Molecular Physiology, Pharmacology, and Therapeutic Implications. Handb Exp Pharmacol 2021; 267:277-356. [PMID: 34345939 DOI: 10.1007/164_2021_501] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
For the past two decades several scholarly reviews have appeared on the inwardly rectifying potassium (Kir) channels. We would like to highlight two efforts in particular, which have provided comprehensive reviews of the literature up to 2010 (Hibino et al., Physiol Rev 90(1):291-366, 2010; Stanfield et al., Rev Physiol Biochem Pharmacol 145:47-179, 2002). In the past decade, great insights into the 3-D atomic resolution structures of Kir channels have begun to provide the molecular basis for their functional properties. More recently, computational studies are beginning to close the time domain gap between in silico dynamic and patch-clamp functional studies. The pharmacology of these channels has also been expanding and the dynamic structural studies provide hope that we are heading toward successful structure-based drug design for this family of K+ channels. In the present review we focus on placing the physiology and pharmacology of this K+ channel family in the context of atomic resolution structures and in providing a glimpse of the promising future of therapeutic opportunities.
Collapse
|
19
|
Bazard P, Frisina RD, Acosta AA, Dasgupta S, Bauer MA, Zhu X, Ding B. Roles of Key Ion Channels and Transport Proteins in Age-Related Hearing Loss. Int J Mol Sci 2021; 22:6158. [PMID: 34200434 PMCID: PMC8201059 DOI: 10.3390/ijms22116158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
The auditory system is a fascinating sensory organ that overall, converts sound signals to electrical signals of the nervous system. Initially, sound energy is converted to mechanical energy via amplification processes in the middle ear, followed by transduction of mechanical movements of the oval window into electrochemical signals in the cochlear hair cells, and finally, neural signals travel to the central auditory system, via the auditory division of the 8th cranial nerve. The majority of people above 60 years have some form of age-related hearing loss, also known as presbycusis. However, the biological mechanisms of presbycusis are complex and not yet fully delineated. In the present article, we highlight ion channels and transport proteins, which are integral for the proper functioning of the auditory system, facilitating the diffusion of various ions across auditory structures for signal transduction and processing. Like most other physiological systems, hearing abilities decline with age, hence, it is imperative to fully understand inner ear aging changes, so ion channel functions should be further investigated in the aging cochlea. In this review article, we discuss key various ion channels in the auditory system and how their functions change with age. Understanding the roles of ion channels in auditory processing could enhance the development of potential biotherapies for age-related hearing loss.
Collapse
Affiliation(s)
- Parveen Bazard
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Robert D. Frisina
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
- Department Communication Sciences and Disorders, College of Behavioral & Communication Sciences, Tampa, FL 33620, USA
| | - Alejandro A. Acosta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Sneha Dasgupta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Mark A. Bauer
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Xiaoxia Zhu
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Bo Ding
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
20
|
Takvam M, Wood CM, Kryvi H, Nilsen TO. Ion Transporters and Osmoregulation in the Kidney of Teleost Fishes as a Function of Salinity. Front Physiol 2021; 12:664588. [PMID: 33967835 PMCID: PMC8098666 DOI: 10.3389/fphys.2021.664588] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Euryhaline teleosts exhibit major changes in renal function as they move between freshwater (FW) and seawater (SW) environments, thus tolerating large fluctuations in salinity. In FW, the kidney excretes large volumes of water through high glomerular filtration rates (GFR) and low tubular reabsorption rates, while actively reabsorbing most ions at high rates. The excreted product has a high urine flow rate (UFR) with a dilute composition. In SW, GFR is greatly reduced, and the tubules reabsorb as much water as possible, while actively secreting divalent ions. The excreted product has a low UFR, and is almost isosmotic to the blood plasma, with Mg2+, SO42–, and Cl– as the major ionic components. Early studies at the organismal level have described these basic patterns, while in the last two decades, studies of regulation at the cell and molecular level have been implemented, though only in a few euryhaline groups (salmonids, eels, tilapias, and fugus). There have been few studies combining the two approaches. The aim of the review is to integrate known aspects of renal physiology (reabsorption and secretion) with more recent advances in molecular water and solute physiology (gene and protein function of transporters). The renal transporters addressed include the subunits of the Na+, K+- ATPase (NKA) enzyme, monovalent ion transporters for Na+, Cl–, and K+ (NKCC1, NKCC2, CLC-K, NCC, ROMK2), water transport pathways [aquaporins (AQP), claudins (CLDN)], and divalent ion transporters for SO42–, Mg2+, and Ca2+ (SLC26A6, SLC26A1, SLC13A1, SLC41A1, CNNM2, CNNM3, NCX1, NCX2, PMCA). For each transport category, we address the current understanding at the molecular level, try to synthesize it with classical knowledge of overall renal function, and highlight knowledge gaps. Future research on the kidney of euryhaline fishes should focus on integrating changes in kidney reabsorption and secretion of ions with changes in transporter function at the cellular and molecular level (gene and protein verification) in different regions of the nephrons. An increased focus on the kidney individually and its functional integration with the other osmoregulatory organs (gills, skin and intestine) in maintaining overall homeostasis will have applied relevance for aquaculture.
Collapse
Affiliation(s)
- Marius Takvam
- Department of Biological Sciences, University of Bergen, Bergen, Norway.,NORCE, Norwegian Research Centre, NORCE Environment, Bergen, Norway
| | - Chris M Wood
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Harald Kryvi
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Tom O Nilsen
- Department of Biological Sciences, University of Bergen, Bergen, Norway.,NORCE, Norwegian Research Centre, NORCE Environment, Bergen, Norway
| |
Collapse
|
21
|
Bamgbola OF, Ahmed Y. Differential diagnosis of perinatal Bartter, Bartter and Gitelman syndromes. Clin Kidney J 2020; 14:36-48. [PMID: 33564404 PMCID: PMC7857843 DOI: 10.1093/ckj/sfaa172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The common finding of hypokalemic alkalosis in several unrelated disorders may confound the early diagnosis of salt-losing tubulopathy (SLT). Antenatal Bartter syndrome (BS) must be considered in idiopathic early-onset polyhydramnios. Fetal megabladder in BS may allow its distinction from third-trimester polyhydramnios that occurs in congenital chloride diarrhea (CCD). Fetal megacolon occurs in CCD while fecal chloride >90 mEq/L in infants is diagnostic. Failure-to-thrive, polydipsia and polyuria in early childhood are the hallmarks of classic BS. Unlike BS, there is low urinary chloride in hypokalemic alkalosis of intractable emesis and cystic fibrosis. Rarely, renal salt wasting may result from cystinosis, Dent disease, disorders of paracellular claudin-10b and Kir4.1 potassium-channel deficiency. Acquired BS may result from calcimimetic up-regulation of a calcium-sensing receptor or autoantibody inactivation of sodium chloride co-transporters in Sjögren syndrome. A relatively common event of heterozygous gene mutations for Gitelman syndrome increases the likelihood of its random occurrence in certain diseases of adult onset. Finally, diuretic abuse is the most common differential diagnosis of SLT. Unlike the persistent elevation in BS, urinary chloride concentration losses waxes and wanes on day-to-day assessment in patients with diuretic misuse.
Collapse
Affiliation(s)
- Oluwatoyin Fatai Bamgbola
- Department of Pediatrics, Division of Pediatric Nephrology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Youssef Ahmed
- Department of Pediatrics, Kings County Hospital, Brooklyn, NY, USA
| |
Collapse
|
22
|
Vega G, Guequén A, Philp AR, Gianotti A, Arzola L, Villalón M, Zegarra-Moran O, Galietta LJ, Mall MA, Flores CA. Lack of Kcnn4 improves mucociliary clearance in muco-obstructive lung disease. JCI Insight 2020; 5:140076. [PMID: 32814712 PMCID: PMC7455130 DOI: 10.1172/jci.insight.140076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Airway mucociliary clearance (MCC) is the main mechanism of lung defense keeping airways free of infection and mucus obstruction. Airway surface liquid volume, ciliary beating, and mucus are central for proper MCC and critically regulated by sodium absorption and anion secretion. Impaired MCC is a key feature of muco-obstructive diseases. The calcium-activated potassium channel KCa.3.1, encoded by Kcnn4, participates in ion secretion, and studies showed that its activation increases Na+ absorption in airway epithelia, suggesting that KCa3.1-induced hyperpolarization was sufficient to drive Na+ absorption. However, its role in airway epithelium is not fully understood. We aimed to elucidate the role of KCa3.1 in MCC using a genetically engineered mouse. KCa3.1 inhibition reduced Na+ absorption in mouse and human airway epithelium. Furthermore, the genetic deletion of Kcnn4 enhanced cilia beating frequency and MCC ex vivo and in vivo. Kcnn4 silencing in the Scnn1b-transgenic mouse (Scnn1btg/+), a model of muco-obstructive lung disease triggered by increased epithelial Na+ absorption, improved MCC, reduced Na+ absorption, and did not change the amount of mucus but did reduce mucus adhesion, neutrophil infiltration, and emphysema. Our data support that KCa3.1 inhibition attenuated muco-obstructive disease in the Scnn1btg/+ mice. K+ channel modulation may be a therapeutic strategy to treat muco-obstructive lung diseases. Silencing the calcium-activated potassium channel KCa.3.1 improves mucociliary clearance in muco-obstructive lung disease by decreasing sodium absorption in the airways.
Collapse
Affiliation(s)
| | - Anita Guequén
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | - Amber R Philp
- Centro de Estudios Científicos, Valdivia, Chile.,Universidad Austral de Chile, Valdivia, Chile
| | | | - Llilian Arzola
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Villalón
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Luis Jv Galietta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Marcus A Mall
- Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research, Berlin, Germany
| | | |
Collapse
|
23
|
Zhang J, Han J, Li L, Zhang Q, Feng Y, Jiang Y, Deng F, Zhang Y, Wu Q, Chen B, Hu J. Inwardly rectifying potassium channel 5.1: Structure, function, and possible roles in diseases. Genes Dis 2020; 8:272-278. [PMID: 33997174 PMCID: PMC8093645 DOI: 10.1016/j.gendis.2020.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/02/2020] [Accepted: 03/13/2020] [Indexed: 11/30/2022] Open
Abstract
Inwardly rectifying potassium (Kir) channels make it easier for K+ to enter into a cell and subsequently regulate cellular biological functions. Kir5.1 (encoded by KCNJ16) alone can form a homotetramer and can form heterotetramers with Kir4.1 (encoded by KCNJ10) or Kir4.2 (encoded by KCNJ15). In most cases, homomeric Kir5.1 is non-functional, while heteromeric Kir5.1 on the cell membrane contributes to the inward flow of K+ ions, which can be regulated by intracellular pH and a variety of signaling mechanisms. In the form of a heterotetramer, Kir5.1 regulates Kir4.1/4.2 activity and is involved in the maintenance of nephron function. Actually, homomeric Kir5.1 may also play a very important role in diseases, including in the ventilatory response to hypoxia and hypercapnia, hearing impairment, cardiovascular disease and cancer. With an increase in the number of studies into the roles of Kir channels, researchers are paying more attention to the pathophysiological functions of Kir5.1. This minireview provides an overview regarding these Kir5.1 roles.
Collapse
Affiliation(s)
- Junhui Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jian Han
- Department of Obstetrics and Gynecology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, PR China
| | - Lingfei Li
- Department of Dermatology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, PR China
| | - Qiong Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yanhai Feng
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Youzhao Jiang
- Department of Endocrinology, People's Hospital of Banan District, Chongqing, 401320, PR China
| | - Fang Deng
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yuping Zhang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Qinan Wu
- Department of Endocrinology, Chongqing Cancer Hospital (Chongqing University Cancer Hospital), Chongqing, 40030, PR China
| | - Bing Chen
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jiongyu Hu
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.,Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| |
Collapse
|
24
|
Li X, Liao J, Jiang Z, Liu X, Chen S, He X, Zhu L, Duan X, Xu Z, Qi B, Guo X, Tong R, Shi J. A concise review of recent advances in anti-heart failure targets and its small molecules inhibitors in recent years. Eur J Med Chem 2020; 186:111852. [PMID: 31759729 DOI: 10.1016/j.ejmech.2019.111852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Heart failure is a disease with high mortality and the risk of heart failure increases in magnitude with age. The patients of heart failure is increasing year by year. Hence, Pharmaceutical researchers have to develop new drugs with better pharmacological effects to coping with this phenomenon. In this article, we reviewed the small molecule compounds for heart failure that have been marketed in recent years or are promising to enter clinical research. We also reviewed the SAR (structure activity relationship) of these molecules, such as renin inhibitors, ROMK inhibitors, a kind of new diuretics, and some dual-targets inhibitors. These small molecules proven to be beneficial effect on heart failure patients. Which may provide ideas for the design of novel anti-heart failure therapeutic drugs.
Collapse
Affiliation(s)
- Xingxing Li
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Jing Liao
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Pediatric Department Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Xinyu Liu
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Shan Chen
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Xia He
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Ling Zhu
- Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Xingmei Duan
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Zhuyu Xu
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Baowen Qi
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Xiaoqiang Guo
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Rongsheng Tong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China.
| | - Jianyou Shi
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China.
| |
Collapse
|
25
|
Defective bicarbonate reabsorption in Kir4.2 potassium channel deficient mice impairs acid-base balance and ammonia excretion. Kidney Int 2019; 97:304-315. [PMID: 31870500 DOI: 10.1016/j.kint.2019.09.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 09/16/2019] [Accepted: 09/23/2019] [Indexed: 11/21/2022]
Abstract
The kidneys excrete the daily acid load mainly by generating and excreting ammonia but the underlying molecular mechanisms are not fully understood. Here we evaluated the role of the inwardly rectifying potassium channel subunit Kir4.2 (Kcnj15 gene product) in this process. In mice, Kir4.2 was present exclusively at the basolateral membrane of proximal tubular cells and disruption of Kcnj15 caused a hyperchloremic metabolic acidosis associated with a reduced threshold for bicarbonate in the absence of a generalized proximal tubule dysfunction. Urinary ammonium excretion rates in Kcnj15- deleted mice were inappropriate to acidosis under basal and acid-loading conditions, and not related to a failure to acidify urine or a reduced expression of ammonia transporters in the collecting duct. In contrast, the expression of key proteins involved in ammonia metabolism and secretion by proximal cells, namely the glutamine transporter SNAT3, the phosphate-dependent glutaminase and phosphoenolpyruvate carboxykinase enzymes, and the sodium-proton exchanger NHE-3 was inappropriate in Kcnj15-deleted mice. Additionally, Kcnj15 deletion depolarized the proximal cell membrane by decreasing the barium-sensitive component of the potassium conductance and caused an intracellular alkalinization. Thus, the Kir4.2 potassium channel subunit is a newly recognized regulator of proximal ammonia metabolism. The kidney consequences of its loss of function in mice support the proposal for KCNJ15 as a molecular basis for human isolated proximal renal tubular acidosis.
Collapse
|
26
|
Galíndez-Cerón JD, Jorge RJB, Chavez-Acosta MH, Jorge ARC, Alves NTQ, Prata MMG, Rodrigues FADP, Havt A, Sampaio TL, Martins AMC, Guerrero-Vargas JA, Monteiro HSA, Beltrán-Vidal JT. Renal Alterations Induced by the Venom of Colombian Scorpion Centruroides Margaritatus. Curr Top Med Chem 2019; 19:2049-2057. [PMID: 31364515 DOI: 10.2174/1568026619666190731143523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Scorpion venom causes renal injury and affects vascular ion-channels function. Centruroides margaritatus scorpion is found in Colombia and is frequently the cause of envenomation accidents; however, its renal impact has never been investigated. OBJECTIVE To evaluate the effects of C. margaritatus venom (CmV) on renal parameters using isolated rat kidney and renal cell culture models. METHODS Wistar rats (n = 5, weighing 240-300 g) were first perfused with Krebs-Henseleit solution containing 6 g 100 mL-1 bovine serum albumin. After 30 minutes, the kidneys were perfused with CmV to a final concentration of 10 μgmL-1; evaluation was performed by measuring Perfusion Pressure (PP), Renal Vascular Resistance (RVR), Urinary Flow (UF), Glomerular Filtration Rate (GFR), and percentage of electrolyte tubular transport. Moreover, kidney histological analyses and cell cytotoxicity in renal tubule epithelial cells (MDCK) and proximal tubular cells (LLC-MK2) were assessed. RESULTS CmV increased PP and RVR 60 min after perfusion. On the other hand, UF, GFR, and the percentages of sodium, potassium and chloride tubular transport decreased after experimental envenomation. UF dropped after 120 min, while GFR and percentage of electrolyte tubular transport diminished after 60, 90 and 120 min. CmV was not toxic to MDCK cell line but reduced the viability of LLC-MK2 cells at concentrations ranging from 6.25 to 200 μgmL-1. Histological analyses disclosed hydropic degeneration, edema, and protein deposits. Flow cytometry disclosed that cell death occurred predominantly by necrosis. CONCLUSION Our results suggest that C. margaritatus venom can trigger renal impairment, mainly in the proximal kidney tubule.
Collapse
Affiliation(s)
- J D Galíndez-Cerón
- Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Grupo de Investigaciones Herpetológicas y Toxinológicas, Universidad del Cauca, Popayán, Colombia
| | - R J B Jorge
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - M H Chavez-Acosta
- Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Grupo de Investigaciones Herpetológicas y Toxinológicas, Universidad del Cauca, Popayán, Colombia
| | - A R C Jorge
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - N T Q Alves
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - M M G Prata
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - F A de Paulo Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - A Havt
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - T L Sampaio
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - A M C Martins
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - J A Guerrero-Vargas
- Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Grupo de Investigaciones Herpetológicas y Toxinológicas, Universidad del Cauca, Popayán, Colombia
| | - H S A Monteiro
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - J T Beltrán-Vidal
- Departamento de Biología, Facultad de Ciencias Naturales, Exactas y de la Educación, Grupo de Investigaciones Herpetológicas y Toxinológicas, Universidad del Cauca, Popayán, Colombia
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Renal ion transport undergoes dramatic changes during the course of gestation. These adaptations are necessary to meet the dynamic requirements of pregnancy and support fetal development. Pregnancy is characterized by a high demand for both sodium and potassium. Recently there has been work in the field profiling the modifications of the renal tubules in pregnancy to meet these demands. The purpose of this review is to summarize these findings. RECENT FINDINGS The work to date suggests an important role for the distal nephron in both the renal sodium and potassium reabsorption during pregnancy. There is strong evidence that renal sodium reabsorption is mediated by the epithelial sodium channel (ENaC). Whereas renal potassium reabsorption is mediated by upregulation of potassium retaining transporters (HKA2) and downregulation of potassium secreting channels (ROMK, BK). SUMMARY Fetal growth restriction and hypertensive disorders of pregnancy including preeclampsia are marked by suboptimal maternal plasma volume expansion, which is determined by renal electrolyte handling. Therefore, understanding the physiologic demand for sodium and potassium in pregnancy and the adaptations required to support these needs is necessary for the effective treatment of diseased states of pregnancy.
Collapse
|
28
|
Ratzan W, Rayaprolu V, Killian SE, Bradley R, Kohout SC. The voltage sensing phosphatase (VSP) localizes to the apical membrane of kidney tubule epithelial cells. PLoS One 2019; 14:e0209056. [PMID: 30964862 PMCID: PMC6456211 DOI: 10.1371/journal.pone.0209056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/21/2019] [Indexed: 12/29/2022] Open
Abstract
Voltage-sensing phosphatases (VSPs) are transmembrane proteins that couple changes in membrane potential to hydrolysis of inositol signaling lipids. VSPs catalyze the dephosphorylation of phosphatidylinositol phosphates (PIPs) that regulate diverse aspects of cell membrane physiology including cell division, growth and migration. VSPs are highly conserved among chordates, and their RNA transcripts have been detected in the adult and embryonic stages of frogs, fish, chickens, mice and humans. However, the subcellular localization and biological function of VSP remains unknown. Using reverse transcriptase-PCR (RT-PCR), we show that both Xenopus laevis VSPs (Xl-VSP1 and Xl-VSP2) mRNAs are expressed in early embryos, suggesting that both Xl-VSPs are involved in early tadpole development. To understand which embryonic tissues express Xl-VSP mRNA, we used in situ hybridization (ISH) and found Xl-VSP mRNA in both the brain and kidney of NF stage 32-36 embryos. By Western blot analysis with a VSP antibody, we show increasing levels of Xl-VSP protein in the developing embryo, and by immunohistochemistry (IHC), we demonstrate that Xl-VSP protein is specifically localized to the apical membrane of both embryonic and adult kidney tubules. We further characterized the catalytic activity of both Xl-VSP homologs and found that while Xl-VSP1 catalyzes 3- and 5-phosphate removal, Xl-VSP2 is a less efficient 3-phosphatase with different substrate specificity. Our results suggest that Xl-VSP1 and Xl-VSP2 serve different functional roles and that VSPs are an integral component of voltage-dependent PIP signaling pathways during vertebrate kidney tubule development and function.
Collapse
Affiliation(s)
- Wil Ratzan
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, United States of America
| | - Vamseedhar Rayaprolu
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, United States of America
| | - Scott E. Killian
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, United States of America
| | - Roger Bradley
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, United States of America
| | - Susy C. Kohout
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana, United States of America
| |
Collapse
|
29
|
Tomilin V, Mamenko M, Zaika O, Wingo CS, Pochynyuk O. TRPV4 deletion protects against hypokalemia during systemic K + deficiency. Am J Physiol Renal Physiol 2019; 316:F948-F956. [PMID: 30838874 DOI: 10.1152/ajprenal.00043.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tight regulation of K+ balance is fundamental for normal physiology. Reduced dietary K+ intake, which is common in Western diets, often leads to hypokalemia and associated cardiovascular- and kidney-related pathologies. The distal nephron, and, specifically, the collecting duct (CD), is the major site of controlled K+ reabsorption via H+-K+-ATPase in the state of dietary K+ deficiency. We (Mamenko MV, Boukelmoune N, Tomilin VN, Zaika OL, Jensen VB, O'Neil RG, Pochynyuk OM. Kidney Int 91: 1398-1409, 2017) have previously demonstrated that the transient receptor potential vanilloid type 4 (TRPV4) Ca2+ channel, abundantly expressed in the CD, contributes to renal K+ handling by promoting flow-induced K+ secretion. Here, we investigated a potential role of TRPV4 in controlling H+-K+-ATPase-dependent K+ reabsorption in the CD. Treatment with a K+-deficient diet (<0.01% K+) for 7 days reduced serum K+ levels in wild-type (WT) mice from 4.3 ± 0.2 to 3.3 ± 0.2 mM but not in TRPV4-/- mice (4.3 ± 0.1 and 4.2 ± 0.3 mM, respectively). Furthermore, we detected a significant reduction in 24-h urinary K+ levels in TRPV4-/- compared with WT mice upon switching to K+-deficient diet. TRPV4-/- animals also had significantly more acidic urine on a low-K+ diet, but not on a regular (0.9% K+) or high-K+ (5% K+) diet, which is consistent with increased H+-K+-ATPase activity. Moreover, we detected a greatly accelerated H+-K+-ATPase-dependent intracellular pH extrusion in freshly isolated CDs from TRPV4-/- compared with WT mice fed a K+-deficient diet. Overall, our results demonstrate a novel kaliuretic role of TRPV4 by inhibiting H+-K+-ATPase-dependent K+ reabsorption in the CD. We propose that TRPV4 inhibition could be a novel strategy to manage certain hypokalemic states in clinical settings.
Collapse
Affiliation(s)
- Viktor Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Transplantation, Department of Medicine, University of Florida , Gainesville, Florida.,North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| |
Collapse
|
30
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
31
|
Nesovic-Ostojic J, Kovacevic S, Spasic S, Lopicic S, Todorovic J, Dincic M, Stanojevic M, Savin M, Milovanovic A, Cemerikic D. Modulation of luminal L-alanine transport in proximal tubular cells of frog kidney induced by low micromolar Cd 2+ concentration. Comp Biochem Physiol C Toxicol Pharmacol 2019; 216:38-42. [PMID: 30414954 DOI: 10.1016/j.cbpc.2018.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/25/2022]
Abstract
The kidneys are recognized as a major target of cadmium-induced toxicity. However, all mechanisms that are involved in the early stages of cadmium nephrotoxicity, particularly considering low micromolar concentrations of cadmium ions (Cd2+) are still unknown. Therefore, the aim of this study was to investigate the effects of peritubular acute exposure to micromolar Cd2+ concentration (2.3 μmol/L) on the rapid depolarization and the rate of slow repolarization of peritubular membrane potential difference (PD), induced by luminal application of L-alanine in proximal tubular cells of frog kidney. The results showed that the luminal application of L-alanine rapidly depolarized the peritubular membrane PD of -42.00 ± 11.68 mV by 23.89 ± 4.15 mV with an average rate of slow repolarization of 5.64 ± 0.81 mV/min. Additionally, peritubular acute exposure to Cd2+ induced change in rapid depolarization of peritubular membrane PD of -53.33 ± 13.01 mV by 18.78 ± 3.31 mV (P < 0.01) after luminal application of L-alanine. Also, peritubular acute exposure to Cd2+ led to statistically significant decrease in the rate of slow repolarization of peritubular membrane PD (3.53 ± 0.35 mV/min; P < 0.05). In conclusion, these results suggest that peritubular acute exposure to low micromolar Cd2+ concentration decreased the rapid depolarization and the rate of slow repolarization of peritubular membrane PD induced by luminal application of L-alanine. This is followed by reduced luminal sodium-coupled transport of L-alanine and this change may be one of the possible mechanisms involved in the early stages of Cd2+-induced nephrotoxicity.
Collapse
Affiliation(s)
| | - Sanjin Kovacevic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Svetolik Spasic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Srdjan Lopicic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Jasna Todorovic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Marko Dincic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Marija Stanojevic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| | - Marina Savin
- Clinic of Nephrology, Clinical Center of Serbia, Medical Faculty, University of Belgrade, Serbia
| | - Aleksandar Milovanovic
- Insitute of Occupational Health, Clinical Center of Serbia, Medical Faculty, University of Belgrade, Serbia
| | - Dusan Cemerikic
- Institute of Pathophysiology, Medical Faculty, University of Belgrade, Serbia
| |
Collapse
|
32
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Zhang G, Gui S, Wang W, Meng D, Meng Q, Luan H, Zhao R, Zhang J, Sui H. Acute stimulatory effect of tumor necrosis factor on the basolateral 50 pS K channels in the thick ascending limb of the rat kidney. Mol Med Rep 2018; 18:4733-4738. [PMID: 30221721 DOI: 10.3892/mmr.2018.9475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 08/13/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the acute effect and mechanism of tumor necrosis factor (TNF) on basolateral 50 pS K channels in the thick ascending limb (TAL) of the rat kidney. The TAL tubules were isolated from the rat kidney, and the activity of the 50 pS K channels was recorded using the patch‑clamp technique. The results indicated that the application of TNF (10 nM) significantly activated the 50 pS K channels and the TNF effect was concentration‑dependent. Inhibition of protein kinase A, phospholipase A2 and protein tyrosine kinase using pathway inhibitors (H89, AACOCF3 and Herbimycin A, respectively) did not abolish the stimulatory effect of TNF, indicating that none of these pathways mediated the TNF effect. By contrast, the phenylarsine oxide inhibitor against protein tyrosine phosphatase (PTP) decreased the activity of the 50 pS K channels and blocked the stimulatory effect of TNF on these channels. Furthermore, western blot analysis demonstrated that the application of TNF (10 nM) in the TAL increased the phosphorylation of PTP, an indication of PTP activity stimulation. Thus, it was concluded that the acute application of TNF may stimulate the basolateral 50 pS K channel in the TAL and the stimulatory effect of TNF may be mediated by the PTP‑dependent pathway.
Collapse
Affiliation(s)
- Guoyan Zhang
- Department of Urology, First Affiliated Hospital, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Shiliang Gui
- Department of Urology, First Affiliated Hospital, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Weiqun Wang
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Dexin Meng
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Qingmin Meng
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Haiyan Luan
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Rixin Zhao
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Jiatian Zhang
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Hongyu Sui
- Department of Physiology, Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| |
Collapse
|
34
|
Palygin O, Pochynyuk O, Staruschenko A. Distal tubule basolateral potassium channels: cellular and molecular mechanisms of regulation. Curr Opin Nephrol Hypertens 2018; 27:373-378. [PMID: 29894319 PMCID: PMC6217967 DOI: 10.1097/mnh.0000000000000437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Multiple clinical and translational evidence support benefits of high potassium diet; however, there many uncertainties underlying the molecular and cellular mechanisms determining effects of dietary potassium. Kir4.1 and Kir5.1 proteins form a functional heteromer (Kir4.1/Kir5.1), which is the primary inwardly rectifying potassium channel on the basolateral membrane of both distal convoluted tubule (DCT) and the collecting duct principal cells. The purpose of this mini-review is to summarize latest advances in our understanding of the evolution, physiological relevance and mechanisms controlling these channels. RECENT FINDINGS Kir4.1 and Kir5.1 channels play a critical role in determining electrolyte homeostasis in the kidney and blood pressure, respectively. It was reported that Kir4.1/Kir5.1 serves as potassium sensors in the distal nephron responding to variations in dietary intake and hormonal stimuli. Global and kidney specific knockouts of either channel resulted in hypokalemia and severe cardiorenal phenotypes. Furthermore, knock out of Kir5.1 in Dahl salt-sensitive rat background revealed the crucial role of the Kir4.1/Kir5.1 channel in salt-induced hypertension. SUMMARY Here, we focus on reviewing novel experimental evidence of the physiological function, expression and hormonal regulation of renal basolateral inwardly rectifying potassium channels. Further investigation of molecular and cellular mechanisms controlling Kir4.1 and Kir4.1/Kir5.1-mediating pathways and development of specific compounds targeting these channels function is essential for proper control of electrolyte homeostasis and blood pressure.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Oleh Pochynyuk
- Department of Integrative Biology, University of Texas Health Science Center, Houston, TX 77030
| | | |
Collapse
|
35
|
Wu P, Gao ZX, Su XT, Ellison DH, Hadchouel J, Teulon J, Wang WH. Role of WNK4 and kidney-specific WNK1 in mediating the effect of high dietary K + intake on ROMK channel in the distal convoluted tubule. Am J Physiol Renal Physiol 2018; 315:F223-F230. [PMID: 29667910 DOI: 10.1152/ajprenal.00050.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
With-no-lysine kinase 4 (WNK4) and kidney-specific (KS)-WNK1 regulate ROMK (Kir1.1) channels in a variety of cell models. We now explore the role of WNK4 and KS-WNK1 in regulating ROMK in the native distal convoluted tubule (DCT)/connecting tubule (CNT) by measuring tertiapin-Q (TPNQ; ROMK inhibitor)-sensitive K+ currents with whole cell recording. TPNQ-sensitive K+ currents in DCT2/CNT of KS- WNK1-/- and WNK4-/- mice were significantly smaller than that of WT mice. In contrast, the basolateral K+ channels (a Kir4.1/5.1 heterotetramer) in the DCT were not inhibited. Moreover, WNK4-/- mice were hypokalemic, while KS- WNK1-/- mice had normal plasma K+ levels. High K+ (HK) intake significantly increased TPNQ-sensitive K+ currents in DCT2/CNT of WT and WNK4-/- mice but not in KS- WNK1-/- mice. However, TPNQ-sensitive K+ currents in the cortical collecting duct (CCD) were normal not only under control conditions but also significantly increased in response to HK in KS- WNK1-/- mice. This suggests that the deletion of KS-WNK1-induced inhibition of ROMK occurs only in the DCT2/CNT. Renal clearance study further demonstrated that the deletion of KS-WNK1 did not affect the renal ability of K+ excretion under control conditions and during increasing K+ intake. Also, HK intake did not cause hyperkalemia in KS- WNK1-/- mice. We conclude that KS-WNK1 but not WNK4 is required for HK intake-induced stimulation of ROMK activity in DCT2/CNT. However, KS-WNK1 is not essential for HK-induced stimulation of ROMK in the CCD, and the lack of KS-WNK1 does not affect net renal K+ excretion.
Collapse
Affiliation(s)
- Peng Wu
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Zhong-Xiuzi Gao
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University , Portland, Oregon
| | - Juliette Hadchouel
- Institut National de la Santé et de la Recherche Médicale, UMR_S1155, University Pierre et Marie Curie, Hospital Tenon , Paris , France.,University Pierre et Marie Curie, Centre de Recherches des Cordeliers, UMR_S1138, Paris , France
| | - Jacques Teulon
- University Pierre et Marie Curie, Centre de Recherches des Cordeliers, UMR_S1138, Paris , France
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
36
|
Inanobe A, Itamochi H, Kurachi Y. Kir Channel Blockages by Proflavine Derivatives via Multiple Modes of Interaction. Mol Pharmacol 2018; 93:592-600. [PMID: 29650538 DOI: 10.1124/mol.117.111377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/06/2018] [Indexed: 11/22/2022] Open
Abstract
Many compounds inhibit tetrameric and pseudo-tetrameric cation channels by associating with the central cavity located in the middle of the membrane plane. They traverse the ion conduction pathway from the intracellular side and through access to the cavity. Previously, we reported that the bacteriostatic agent, proflavine, preferentially blocked a subset of inward rectifier K+ (Kir) channels. However, the development of the inhibition of Kir1.1 by the compound was obviously different from that operating in Kir3.2 as a pore blocker. To gain mechanistic insights into the compound-channel interaction, we analyzed its chemical specificity, subunit selectivity, and voltage dependency using 13 different combinations of Kir-channel family members and 11 proflavine derivatives. The Kir-channel family members were classified into three groups: 1) Kir2.2, Kir3.x, Kir4.2, and Kir6.2Δ36, which exhibited Kir3.2-type inhibition (slow onset and recovery, irreversible, and voltage-dependent blockage); 2) Kir1.1 and Kir4.1/Kir5.1 (prompt onset and recovery, reversible, and voltage-independent blockage); and 3) Kir2.1, Kir2.3, Kir4.1, and Kir7.1 (no response). The degree of current inhibition depended on the combination of compounds and channels. Chimera between proflavine-sensitive Kir1.1 and -insensitive Kir4.1 revealed that the extracellular portion of Kir1.1 is crucial for the recognition of the proflavine derivative acrinol. In conclusion, preferential blockage of Kir-channel family members by proflavine derivatives is based on multiple modes of action. This raises the possibility of designing subunit-specific inhibitors.
Collapse
Affiliation(s)
- Atsushi Inanobe
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| | - Hideaki Itamochi
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| | - Yoshihisa Kurachi
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
37
|
Saidani C, Béchohra L, Laraba-Djebari F, Hammoudi-Triki D. Kidney inflammation and tissue injury induced by scorpion venom: comparison with a nephrotoxic model. TOXIN REV 2018. [DOI: 10.1080/15569543.2018.1446028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Chanez Saidani
- Laboratory of Cellular and Molecular Biology, University of Science and Technology Houari Boumediene, Algiers, Algeria
| | - Louisa Béchohra
- Laboratory of Cellular and Molecular Biology, University of Science and Technology Houari Boumediene, Algiers, Algeria
| | - Fatima Laraba-Djebari
- Laboratory of Cellular and Molecular Biology, University of Science and Technology Houari Boumediene, Algiers, Algeria
| | - Djelila Hammoudi-Triki
- Laboratory of Cellular and Molecular Biology, University of Science and Technology Houari Boumediene, Algiers, Algeria
| |
Collapse
|
38
|
West CA, Welling PA, West DA, Coleman RA, Cheng KY, Chen C, DuBose TD, Verlander JW, Baylis C, Gumz ML. Renal and colonic potassium transporters in the pregnant rat. Am J Physiol Renal Physiol 2018; 314:F251-F259. [PMID: 29046297 PMCID: PMC5866449 DOI: 10.1152/ajprenal.00288.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 11/22/2022] Open
Abstract
Gestational potassium retention, most of which occurs during late pregnancy, is essential for fetal development. The purpose of this study was to examine mechanisms underlying changes in potassium handling by the kidney and colon in pregnancy. We found that potassium intake and renal excretion increased in late pregnancy while fecal potassium excretion remained unchanged and that pregnant rats exhibited net potassium retention. By quantitative PCR we found markedly increased H+-K+-ATPase type 2 (HKA2) mRNA expression in the cortex and outer medullary of late pregnant vs. virgin. Renal outer medullary potassium channel (ROMK) mRNA was unchanged in the cortex, but apical ROMK abundance (by immunofluorescence) was decreased in pregnant vs. virgin in the distal convoluted tubule (DCT) and connecting tubule (CNT). Big potassium-α (BKα) channel-α protein abundance in intercalated cells in the cortex and outer medullary collecting ducts (by immunohistochemistry) fell in late pregnancy. In the distal colon we found increased HKA2 mRNA and protein abundance (Western blot) and decreased BKα protein with no observed changes in mRNA. Therefore, the potassium retention of pregnancy is likely to be due to increased collecting duct potassium reabsorption (via increased HKA2), decreased potassium secretion (via decreased ROMK and BK), as well as increased colonic reabsorption via HKA2.
Collapse
Affiliation(s)
- Crystal A West
- Department of Medicine, Georgetown University, Washington, District of Columiba
| | - Paul A Welling
- Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| | - David A West
- Department of Medicine, Georgetown University, Washington, District of Columiba
| | - Richard A Coleman
- Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Kit-Yan Cheng
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Chao Chen
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Thomas D DuBose
- Department of Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Jill W Verlander
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Chris Baylis
- Department of Medicine, University of Florida , Gainesville, Florida
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Michelle L Gumz
- Department of Medicine, University of Florida , Gainesville, Florida
| |
Collapse
|
39
|
Huang X, Lee SH, Lu H, Sanders KM, Koh SD. Molecular and functional characterization of inwardly rectifying K + currents in murine proximal colon. J Physiol 2018; 596:379-391. [PMID: 29205356 PMCID: PMC5792581 DOI: 10.1113/jp275234] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Interstitial cells of Cajal (ICC) from murine colonic muscles express genes encoding inwardly rectifying K+ channels. Transcripts of Kcnj2 (Kir2.1), Kcnj4 (Kir2.3), Kcnj14 (Kir2.4), Kcnj5 (Kir3.4), Kcnj8 (Kir 6.1) and Kcnj11 (Kir6.2) were found in colonic ICC. A conductance with properties consistent with Kir2 channels was observed in ICC but not in smooth muscle cells (SMC). Despite expression of gene transcripts, G-protein gated K+ channel (Kir3) and KATP (Kir6) currents were not resolved in ICC. KATP is a conductance prominent in SMC. Kir2 antagonist caused depolarization of freshly dispersed ICC and colonic smooth muscles, suggesting that this conductance is active under resting conditions in colonic muscles. The conclusion of the present study is that ICC express the Ba2+ -sensitive, inwardly rectifying K+ conductance in colonic muscles. This conductance is most probably a result of heterotetramers of Kir2 gene products, with this regulating resting potentials and the excitability of colonic muscles. ABSTRACT Membrane potentials of gastrointestinal muscles are important because voltage-dependent Ca2+ channels in smooth muscle cells (SMC) provide the Ca2+ that triggers contraction. Regulation of membrane potential is complicated because SMC are electrically coupled to interstitial cells of Cajal (ICC) and PDGFRα+ cells. Activation of conductances in any of these cells affects the excitability of the syncytium. We explored the role of inward rectifier K+ conductances in colonic ICC that might contribute to regulation of membrane potential. ICC expressed Kcnj2 (Kir2.1), Kcnj4 (Kir2.3), Kcnj14 (Kir2.4), Kcnj5 (Kir3.4), Kcnj8 (Kir 6.1) and Kcnj11 (Kir6.2). Voltage clamp experiments showed activation of inward current when extracellular K+ ([K+ ]o ) was increased. The current was inwardly rectifying and inhibited by Ba2+ (10 μm) and ML-133 (10 μm). A similar current was not available in SMC. The current activated in ICC by elevated [K+ ]o was not affected by Tertiapin-Q. Gβγ, when dialysed into cells, failed to activate a unique, Tertiapin-Q-sensitive conductance. Freshly dispersed ICC showed no evidence of functional KATP . Pinacidil failed to activate current and the inward current activated by elevated [K+ ]o was insensitive to glibenclamide. Under current clamp, ML-133 caused the depolarization of isolated ICC and also that of cells impaled with microelectrodes in intact muscle strips. These findings show that ICC, when isolated freshly from colonic muscles, expressed a Ba2+ -sensitive, inwardly rectifying K+ conductance. This conductance is most probably a result of the expression of multiple Kir2 family paralogues, and the inwardly rectifying conductance contributes to the regulation of resting potentials and excitability of colonic muscles.
Collapse
Affiliation(s)
- Xu Huang
- Department of Physiology and Cell BiologyUniversity of Nevada School of MedicineRenoNVUSA
| | - Si Hyung Lee
- Present address: Division of Gastroenterology and Hepatology, Department of Internal MedicineYeungnam University College of MedicineNam‐GuDaeguSouth Korea
| | - Hongli Lu
- Department of Physiology and Cell BiologyUniversity of Nevada School of MedicineRenoNVUSA
| | - Kenton M. Sanders
- Department of Physiology and Cell BiologyUniversity of Nevada School of MedicineRenoNVUSA
| | - Sang Don Koh
- Department of Physiology and Cell BiologyUniversity of Nevada School of MedicineRenoNVUSA
| |
Collapse
|
40
|
Nanazashvili M, Sánchez-Rodríguez JE, Fosque B, Bezanilla F, Sackin H. LRET Determination of Molecular Distances during pH Gating of the Mammalian Inward Rectifier Kir1.1b. Biophys J 2018; 114:88-97. [PMID: 29320699 PMCID: PMC5773755 DOI: 10.1016/j.bpj.2017.10.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/23/2017] [Accepted: 10/19/2017] [Indexed: 01/31/2023] Open
Abstract
Gating of the mammalian inward rectifier Kir1.1 at the helix bundle crossing (HBC) by intracellular pH is believed to be mediated by conformational changes in the C-terminal domain (CTD). However, the exact motion of the CTD during Kir gating remains controversial. Crystal structures and single-molecule fluorescence resonance energy transfer of KirBac channels have implied a rigid body rotation and/or a contraction of the CTD as possible triggers for opening of the HBC gate. In our study, we used lanthanide-based resonance energy transfer on single-Cys dimeric constructs of the mammalian renal inward rectifier, Kir1.1b, incorporated into anionic liposomes plus PIP2, to determine unambiguous, state-dependent distances between paired Cys residues on diagonally opposite subunits. Functionality and pH dependence of our proteoliposome channels were verified in separate electrophysiological experiments. The lanthanide-based resonance energy transfer distances measured in closed (pH 6) and open (pH 8) conditions indicated neither expansion nor contraction of the CTD during gating, whereas the HBC gate widened by 8.8 ± 4 Å, from 6.3 ± 2 to 15.1 ± 6 Å, during opening. These results are consistent with a Kir gating model in which rigid body rotation of the large CTD around the permeation axis is correlated with opening of the HBC hydrophobic gate, allowing permeation of a 7 Å hydrated K ion.
Collapse
Affiliation(s)
- Mikheil Nanazashvili
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois
| | - Jorge E Sánchez-Rodríguez
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois; Departamento de Física, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ben Fosque
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
| | - Henry Sackin
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois.
| |
Collapse
|
41
|
Kim JM, Xu S, Guo X, Hu H, Dong K, Wang T. Urinary bladder hypertrophy characteristic of male ROMK Bartter's mice does not occur in female mice. Am J Physiol Regul Integr Comp Physiol 2017; 314:R334-R341. [PMID: 29092859 DOI: 10.1152/ajpregu.00315.2017] [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] [Indexed: 01/16/2023]
Abstract
The renal outer medullary potassium channel (ROMK; Kir1.1) plays an important role in Na+ and K+ homeostasis. ROMK knockout (KO) mice show a similar phenotype to Bartter's syndrome of salt wasting and dehydration due to reduced Na-2Cl-K-cotransporter activity but not in ROMK1 KO mice. ROMK KO mice also show hydronephrosis; however, the mechanism of this phenotype has not been understood. We have previously demonstrated a gender-sex difference in hydronephrosis and PGE2 production in ROMK KO mice. In this study we compared the gender-sex difference in bladder hypertrophy and hydronephrosis in ROMK KO mice. The bladder weight, bladder capacity, and the thickness of urothelium in male ROMK KO showed average increased two to approximately fourfold greater than wild-type (WT) mice, but there was no difference in either female or ROMK1 KO mice. The thickness of the urothelium was 648.8 ± 33.2 µm vs. 302.7 ± 16.5 µm ( P < 0.001) and the detrusor muscle 1,940.7 ± 98.9 µm vs. 1,308.2 ± 102.1 µm ( P = 0.013), respectively, in 12-mo male ROMK KO mice compared with the same age WT mice. Western blotting detected ROMK expression at 45~48 kDa, and both ROMK1 and ROMK2 mRNA were detected by quantitative PCR in the bladder. Immunofluorescence staining showed ROMK stained in the bladder, ureter, and urethra in WT but not in KO. In addition, there was a correlation between the severity of hydronephrosis and the bladder weight in male but not in female ROMK KO mice. In conclusion, ROMK expressed in the urinary tract at both protein and mRNA levels; significant enlargement and hypertrophy of the bladder may contribute to hydronephrosis in male ROMK KO mice.
Collapse
Affiliation(s)
- Jun-Mo Kim
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| | - Shuhua Xu
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| | - Xiaoyun Guo
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| | - Haiyan Hu
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| | - Ke Dong
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| | - Tong Wang
- Department of Cellular and Molecular Physiology, Yale University, School of Medicine , New Haven, Connecticut
| |
Collapse
|
42
|
|
43
|
Wang B, Wen D, Li H, Wang-France J, Sansom SC. Net K + secretion in the thick ascending limb of mice on a low-Na, high-K diet. Kidney Int 2017; 92:864-875. [PMID: 28688582 DOI: 10.1016/j.kint.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/23/2017] [Accepted: 04/06/2017] [Indexed: 12/29/2022]
Abstract
Because of its cardio-protective effects, a low-Na, high-K diet (LNaHK) is often warranted in conjunction with diuretics to treat hypertensive patients. However, it is necessary to understand the renal handling of such diets in order to choose the best diuretic. Wild-type (WT) or Renal Outer Medullary K channel (ROMK) knockout mice (KO) were given a regular (CTRL), LNaHK, or high-K diet (HK) for 4-7 days. On LNaHK, mice treated with either IP furosemide for 12 hrs, or given furosemide in drinking water for 7 days, exhibited decreased K clearance. We used free-flow micropuncture to measure the [K+] in the early distal tubule (EDT [K+]) before and after furosemide treatment. Furosemide increased the EDT [K+] in WT on CTRL but decreased that in WT on LNaHK. Furosemide did not affect the EDT [K+] of KO on LNaHK or WT on HK. Furosemide-sensitive Na+ excretion was significantly greater in mice on LNaHK than those on CTRL or HK. Patch clamp analysis of split-open TALs revealed that 70-pS ROMK exhibited a higher open probability (Po) but similar density in mice on LNaHK, compared with CTRL. No difference was found in the density or Po of the 30 pS K channels between the two groups. These results indicate mice on LNaHK exhibited furosemide-sensitive net K+ secretion in the TAL that is dependent on increased NKCC2 activity and mediated by ROMK. We conclude that furosemide is a K-sparing diuretic by decreasing the TAL net K+ secretion in subjects on LNaHK.
Collapse
Affiliation(s)
- Bangchen Wang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Donghai Wen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Huaqing Li
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jun Wang-France
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steven C Sansom
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
44
|
Shibata S. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: Mineralocorticoid receptor and NaCl transport mechanisms in the renal distal nephron. J Endocrinol 2017; 234:T35-T47. [PMID: 28341694 DOI: 10.1530/joe-16-0669] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023]
Abstract
A key role of aldosterone and mineralocorticoid receptor is to regulate fluid volume and K+ homeostasis in the body by acting on the renal distal nephron. Global responses of the kidney to elevated aldosterone levels are determined by the coordinate action of different constituent tubule cells, including principal cells, intercalated cells and distal convoluted tubule cells. Recent studies on genetic mutations causing aldosterone overproduction have identified the molecules involved in aldosterone biosynthesis in the adrenal gland, and there is also increasing evidence for mechanisms and signaling pathways regulating the balance between renal NaCl reabsorption and K+ secretion, the two major effects of aldosterone. In particular, recent studies have demonstrated that mineralocorticoid receptor in intercalated cells is selectively regulated by phosphorylation, which prevents ligand binding and activation. Moreover, the ubiquitin ligase complex composed of Kelch-like 3 and Cullin 3 acts downstream of angiotensin II and plasma K+ alterations, regulating Na-Cl cotransporter independently of aldosterone in distal convoluted tubule cells. These and other effects are integrated to produce appropriate kidney responses in a high-aldosterone state, and are implicated in fluid and electrolyte disorders in humans. This review summarizes the current knowledge on mechanisms modulating mineralocorticoid receptor and its downstream effectors in the distal nephron.
Collapse
Affiliation(s)
- Shigeru Shibata
- Division of NephrologyDepartment of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
- Division of Clinical EpigeneticsResearch Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Tachow A, Thoungseabyoun W, Phuapittayalert L, Petcharat K, Sakagami H, Kondo H, Hipkaeo W. Co-localization of endogenous Arf6 and its activator EFA6D in the granular convoluted tubule cells of mouse submandibular glands under normal conditions and when stimulated by isoproterenol, noradrenaline and carbachol. Arch Oral Biol 2017. [PMID: 28645101 DOI: 10.1016/j.archoralbio.2017.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study proposed to investigate the localization at light and electron microscopic levels of Arf6 and its activator EFA6D in the mouse submandibular gland (SMG) under normal conditions and when stimulated by adrenergic or cholinergic agonists. MATERIALS AND METHODS SMGs of male adult mice were utilized for immunoblotting and immuno-light and -electron microscopic analyses. Isoproterenol and noradrenalin were used as adrenergics, while carbachol was used for the cholinergic stimulant. SMGs were examined at 15, 30, 60 and 120min after intraperitoneal injection of these agents. RESULTS Immunoreactivities for both Arf6 and its activator EFA6D were similarly intense in the basolateral domain of GCTs, but no significant immunoreactivities were seen in the apical domain of GCT cells or any domain of acinar cells under normal conditions. In immuno-electron microscopy, the immunoreactive materials were mainly deposited on the basolateral plasma membranes and subjacent cytoplasm. Shortly after injection of isoproterenol and noradrenaline, but not carbachol, the immunoreactivities for both molecules were additionally seen on the apical plasmalemma of most, if not all, GCT cells, but not acinar cells. CONCLUSION The present findings suggest that the direct involvement of Arf6/EFA6D in regulatory exocytosis at the apical plasma membrane of acinar and GCT cells is apparently to be smaller, if present, than that of endocytosis at the basolateral membranes of GCT cells under normal conditions. This also suggests that the two molecules function additionally at the apical membrane of GCT cells for modulation of saliva secretion under β-adrenoceptor stimulation.
Collapse
Affiliation(s)
- Apussara Tachow
- Nanomorphology-Based Apply Research Group & Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Wipawee Thoungseabyoun
- Nanomorphology-Based Apply Research Group & Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Kanoktip Petcharat
- Biochemistry and Nutrition, School of Medical Science, University of Phayao, Phayao, Thailand
| | - Hiroyuki Sakagami
- Department of Anatomy, School of Medicine, Kitasato University, Tokyo, Japan
| | - Hisatake Kondo
- Nanomorphology-Based Apply Research Group & Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Wiphawi Hipkaeo
- Nanomorphology-Based Apply Research Group & Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
46
|
O'Donnell BM, Mackie TD, Subramanya AR, Brodsky JL. Endoplasmic reticulum-associated degradation of the renal potassium channel, ROMK, leads to type II Bartter syndrome. J Biol Chem 2017. [PMID: 28630040 DOI: 10.1074/jbc.m117.786376] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type II Bartter syndrome is caused by mutations in the renal outer medullary potassium (ROMK) channel, but the molecular mechanisms underlying this disease are poorly defined. To rapidly screen for ROMK function, we developed a yeast expression system and discovered that yeast cells lacking endogenous potassium channels could be rescued by WT ROMK but not by ROMK proteins containing any one of four Bartter mutations. We also found that the mutant proteins were significantly less stable than WT ROMK. However, their degradation was slowed in the presence of a proteasome inhibitor or when yeast cells contained mutations in the CDC48 or SSA1 gene, which is required for endoplasmic reticulum (ER)-associated degradation (ERAD). Consistent with these data, sucrose gradient centrifugation and indirect immunofluorescence microscopy indicated that most ROMK protein was ER-localized. To translate these findings to a more relevant cell type, we measured the stabilities of WT ROMK and the ROMK Bartter mutants in HEK293 cells. As in yeast, the Bartter mutant proteins were less stable than the WT protein, and their degradation was slowed in the presence of a proteasome inhibitor. Finally, we discovered that low-temperature incubation increased the steady-state levels of a Bartter mutant, suggesting that the disease-causing mutation traps the protein in a folding-deficient conformation. These findings indicate that the underlying pathology for at least a subset of patients with type II Bartter syndrome is linked to the ERAD pathway and that future therapeutic strategies should focus on correcting deficiencies in ROMK folding.
Collapse
Affiliation(s)
- Brighid M O'Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
47
|
Improvement of hERG-ROMK index of spirocyclic ROMK inhibitors through scaffold optimization and incorporation of novel pharmacophores. Bioorg Med Chem Lett 2017; 27:2559-2566. [DOI: 10.1016/j.bmcl.2017.03.086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/26/2017] [Accepted: 03/28/2017] [Indexed: 12/15/2022]
|
48
|
Wang L, Zhang C, Su XT, Lin DH, Wu P, Schwartzman ML, Wang WH. PGF 2α regulates the basolateral K channels in the distal convoluted tubule. Am J Physiol Renal Physiol 2017; 313:F254-F261. [PMID: 28356287 PMCID: PMC5582901 DOI: 10.1152/ajprenal.00102.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 11/22/2022] Open
Abstract
Our aim is to examine the role of PGF2α receptor (FP), a highly expressed prostaglandin receptor in the distal convoluted tubule (DCT) in regulating the basolateral 40-pS K channel. The single-channel studies demonstrated that PGF2α had a biphasic effect on the 40-pS K channel in the DCT-PGF2α stimulated at low concentrations (less than 500 nM), while at high concentrations (above 1 µM), it inhibited the 40-pS K channels. Moreover, neither 13,14-dihydro-15-keto-PGF2α (a metabolite of PGF2α) nor PGE2 was able to mimic the effect of PGF2α on the 40-pS K channel in the DCT. The inhibition of PKC had no significant effect on the 40-pS K channel; however, it abrogated the inhibitory effect of 5 µM PGF2α on the K channel. Moreover, stimulation of PKC inhibited the 40-pS K channel in the DCT, suggesting that PKC mediates the inhibitory effect of PGF2α on the 40-pS K channel. Conversely, the stimulatory effect of PGF2α on the 40-pS K channel was absent in the DCT treated with DPI, a NADPH oxidase (NOX) inhibitor. Also, adding 100 µM H2O2 mimicked the stimulatory effect of PGF2α and increased the 40-pS K channel activity in DCT. Moreover, the stimulatory effect of 500 nM PGF2α and H2O2 was not additive, suggesting the role of superoxide-related species in mediating the stimulatory effect of PGF2α on the 40-pS K channel. The inhibition of Src family tyrosine protein kinase (SFK) not only inhibited the 40-pS K channel in the DCT but also completely abolished the stimulatory effects of PGF2α and H2O2 on the 40-pS K channel. We conclude that PGF2α at low doses stimulates the basolateral 40-pS K channel by a NOX- and SFK-dependent mechanism, while at high concentrations, it inhibits the K channel by a PKC-dependent pathway.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Physiology, Harbin Medical University, Harbin, China; and.,Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Chengbiao Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Peng Wu
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | | | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
49
|
Lee JD, Lee MH, Yang WK, Wang KL, Lee TH. Differential Expression of Renal Outer Medullary K + Channel and Voltage-gated K + Channel 7.1 in Bladder Urothelium of Patients With Interstitial Cystitis/Painful Bladder Syndrome. Urology 2017; 101:169.e1-169.e5. [DOI: 10.1016/j.urology.2016.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 10/04/2016] [Accepted: 11/04/2016] [Indexed: 12/27/2022]
|
50
|
McDonough AA, Youn JH. Potassium Homeostasis: The Knowns, the Unknowns, and the Health Benefits. Physiology (Bethesda) 2017; 32:100-111. [PMID: 28202621 PMCID: PMC5337831 DOI: 10.1152/physiol.00022.2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Potassium homeostasis has a very high priority because of its importance for membrane potential. Although extracellular K+ is only 2% of total body K+, our physiology was evolutionarily tuned for a high-K+, low-Na+ diet. We review how multiple systems interface to accomplish fine K+ balance and the consequences for health and disease.
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Jang H Youn
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|