1
|
Kok M, Singh I, Aizenman E, Brodsky JL. Inefficient maturation of disease-linked mutant forms of the KCC2 potassium-chloride cotransporter correlates with predicted pathogenicity. J Biol Chem 2025; 301:108399. [PMID: 40074080 PMCID: PMC12001125 DOI: 10.1016/j.jbc.2025.108399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The potassium-chloride cotransporter 2 (KCC2) is required for neuronal development, and KCC2 dysregulation is implicated in several neurodevelopmental disorders, including schizophrenia, autism, and epilepsy. A dozen mutations in the KCC2-encoding gene, SLC12A5, are associated with these disorders, but few are fully characterized. To this end, we examined KCC2 biogenesis in a HEK293 cell model. While most of the examined disease-associated mutants matured efficiently, the L403P mutant was unable to traffic to the Golgi. Two other mutants, A191V and R857L, exhibited more subtle defects in maturation. Cell surface biotinylation assays showed that these mutants were also depleted from the cell surface. Another disease-associated variant, R952H, acquired Golgi-associated glycans yet was significantly depleted from the plasma membrane, consistent with loss of a plasma membrane-stabilizing phosphorylation site. To determine whether the ability of KCC2 to mature to the Golgi could be predicted, we employed a computational pathogenicity program, Rhapsody, which was shown in past work to predict endoplasmic reticulum-associated degradation-targeting of an unrelated ion channel. We discovered that the Rhapsody pathogenicity score correlated with relative defects in KCC2 maturation, and the algorithm outperformed two other commonly used programs. These data demonstrate the efficacy of a bioinformatic tool to predict the efficiency of KCC2 biogenesis. We also propose that Rhapsody can be used to develop hypotheses on defects associated with other disease-associated SLC12A5 alleles as they are identified.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ishika Singh
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
2
|
Busselman BW, Ratnayake I, Terasaki MR, Thakkar VP, Ilyas A, Otterpohl KL, Zimmerman JL, Chandrasekar I. Actin cytoskeleton and associated myosin motors within the renal epithelium. Am J Physiol Renal Physiol 2024; 327:F553-F565. [PMID: 39052845 PMCID: PMC11483076 DOI: 10.1152/ajprenal.00078.2024] [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: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.
Collapse
Affiliation(s)
- Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | | | - Mark R Terasaki
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| |
Collapse
|
3
|
Lasaad S, Nickerson AJ, Crambert G, Satlin LM, Kleyman TR. Going with the flow: New insights regarding flow induced K + secretion in the distal nephron. Physiol Rep 2024; 12:e70087. [PMID: 39428258 PMCID: PMC11491169 DOI: 10.14814/phy2.70087] [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: 09/12/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
K+ secretion in the distal nephron has a critical role in K+ homeostasis and is the primary route by which K+ is lost from the body. Renal K+ secretion is enhanced by increases in dietary K+ intake and by increases in tubular flow rate in the distal nephron. This review addresses new and important insights regarding the mechanisms underlying flow-induced K+ secretion (FIKS). While basal K+ secretion in the distal nephron is mediated by renal outer medullary K+ (ROMK) channels in principal cells (PCs), FIKS is mediated by large conductance, Ca2+/stretch activated K+ (BK) channels in intercalated cells (ICs), a distinct cell type. BK channel activation requires an increase in intracellular Ca2+ concentration ([Ca2+]i), and both PCs and ICs exhibit increases in [Ca2+]i in response to increases in tubular fluid flow rate, associated with an increase in tubular diameter. PIEZO1, a mechanosensitive, nonselective cation channel, is expressed in the basolateral membranes of PCs and ICs, where it functions as a mechanosensor. The loss of flow-induced [Ca2+]i transients in ICs and BK channel-mediated FIKS in microperfused collecting ducts isolated from mice with IC-specific deletion of Piezo1 in the CCD underscores the importance of PIEZO1 in the renal regulation of K+ transport.
Collapse
Affiliation(s)
- Samia Lasaad
- Department of PediatricsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Gilles Crambert
- Centre de Recherche Des Cordeliers, Institut National de la Santé et de la Recherche Scientifique (INSERM)Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et TubulopathiesParisFrance
- Unité Métabolisme et Physiologie RénaleCentre National de la Recherche Scientifique (CNRS) EMR 8228ParisFrance
| | - Lisa M. Satlin
- Department of PediatricsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Thomas R. Kleyman
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Cell Biology and Department of Pharmacology and Chemical BiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
4
|
Guo J, Zhang C, Zhao H, Yan Y, Liu Z. The key mediator of diabetic kidney disease: Potassium channel dysfunction. Genes Dis 2024; 11:101119. [PMID: 38523672 PMCID: PMC10958065 DOI: 10.1016/j.gendis.2023.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 06/11/2022] [Accepted: 06/04/2023] [Indexed: 03/26/2024] Open
Abstract
Diabetic kidney disease is a leading cause of end-stage renal disease, making it a global public health concern. The molecular mechanisms underlying diabetic kidney disease have not been elucidated due to its complex pathogenesis. Thus, exploring these mechanisms from new perspectives is the current focus of research concerning diabetic kidney disease. Ion channels are important proteins that maintain the physiological functions of cells and organs. Among ion channels, potassium channels stand out, because they are the most common and important channels on eukaryotic cell surfaces and function as the basis for cell excitability. Certain potassium channel abnormalities have been found to be closely related to diabetic kidney disease progression and genetic susceptibility, such as KATP, KCa, Kir, and KV. In this review, we summarized the roles of different types of potassium channels in the occurrence and development of diabetic kidney disease to discuss whether the development of DKD is due to potassium channel dysfunction and present new ideas for the treatment of DKD.
Collapse
Affiliation(s)
- Jia Guo
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan 450052, China
- Research Center for Kidney Disease, Zhengzhou, Henan 450052, China
| | - Chaojie Zhang
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan 450052, China
- Research Center for Kidney Disease, Zhengzhou, Henan 450052, China
| | - Hui Zhao
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan 450052, China
- Research Center for Kidney Disease, Zhengzhou, Henan 450052, China
| | - Yufan Yan
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan 450052, China
- Research Center for Kidney Disease, Zhengzhou, Henan 450052, China
| | - Zhangsuo Liu
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan 450052, China
- Research Center for Kidney Disease, Zhengzhou, Henan 450052, China
| |
Collapse
|
5
|
Abstract
This interdisciplinary review explores the intricate nexus between HIV infection, nutrition, adrenal gland function, and cardiovascular health, highlighting a critical aspect of HIV management often overlooked in current literature. With the advent of antiretroviral therapy, the life expectancy of people living with HIV has dramatically improved, transforming HIV into a manageable chronic condition. However, this success brings forth new challenges, notably an increased risk of cardiovascular diseases among people living with HIV. We examine the normal physiology of the adrenal gland, including its role in mineral metabolism, a crucial facet of nutrition. We discuss the evolution of knowledge tying adrenal pathology to cardiovascular disease. We explore the impact of HIV on adrenal gland findings from a gross pathology perspective, as well as the clinical impact of adrenal insufficiency in HIV. The review further elucidates the role of nutrition in this context, considering the double burden of undernutrition and obesity prevalent in regions heavily affected by HIV. By aggregating findings from longitudinal studies and recent clinical trials, the review presents compelling evidence of increased cardiovascular disease among people living with HIV compared with people without HIV. It highlights the critical role of the adrenal glands in regulating nutrient metabolism and its implications for cardiovascular health, drawing attention to the potential for dietary interventions and targeted therapies to mitigate these risks. This review urges a paradigm shift in the management of HIV, advocating for a holistic approach that incorporates nutritional assessment and interventions into routine HIV care to address the complex interplay between HIV, adrenal function, and cardiovascular health. Through this lens, we offer insights into novel therapeutic strategies aimed at reducing cardiovascular risk in people living with HIV, contributing to the ongoing efforts to enhance the quality of life and longevity in this population.
Collapse
Affiliation(s)
- Anxious J Niwaha
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Uganda Research Unit, Entebbe (A.J.N.)
| | - James Brian Byrd
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (J.B.B.)
| |
Collapse
|
6
|
Welling PA, Little R, Al-Qusairi L, Delpire E, Ellison DH, Fenton RA, Grimm PR. Potassium-Switch Signaling Pathway Dictates Acute Blood Pressure Response to Dietary Potassium. Hypertension 2024; 81:1044-1054. [PMID: 38465625 PMCID: PMC11023808 DOI: 10.1161/hypertensionaha.123.22546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Potassium (K+)-deficient diets, typical of modern processed foods, increase blood pressure (BP) and NaCl sensitivity. A K+-dependent signaling pathway in the kidney distal convoluted tubule, coined the K+ switch, that couples extracellular K+ sensing to activation of the thiazide-sensitive NaCl cotransporter (NCC) and NaCl retention has been implicated, but causality has not been established. METHODS To test the hypothesis that small, physiological changes in plasma K+ (PK+) are translated to BP through the switch pathway, a genetic approach was used to activate the downstream switch kinase, SPAK (SPS1-related proline/alanine-rich kinase), within the distal convoluted tubule. The CA-SPAK (constitutively active SPS1-related proline/alanine-rich kinase mice) were compared with control mice over a 4-day PK+ titration (3.8-5.1 mmol) induced by changes in dietary K+. Arterial BP was monitored using radiotelemetry, and renal function measurements, NCC abundance, phosphorylation, and activity were made. RESULTS As PK+ decreased in control mice, BP progressively increased and became sensitive to dietary NaCl and hydrochlorothiazide, coincident with increased NCC phosphorylation and urinary sodium retention. By contrast, BP in CA-SPAK mice was elevated, resistant to the PK+ titration, and sensitive to hydrochlorothiazide and salt at all PK+ levels, concomitant with sustained and elevated urinary sodium retention and NCC phosphorylation and activity. Thus, genetically locking the switch on drives NaCl sensitivity and prevents the response of BP to potassium. CONCLUSIONS Low K+, common in modern ultraprocessed diets, presses the K+-switch pathway to turn on NCC activity, increasing sodium retention, BP, and salt sensitivity.
Collapse
Affiliation(s)
- Paul A. Welling
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Robert Little
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Lama Al-Qusairi
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, USA
| | - David H. Ellison
- Department of Medicine, Division of Nephrology, Oregon Health Science Center, Portland, Oregon, US
| | - Robert A. Fenton
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - P. Richard Grimm
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
7
|
Nie J, Yang H, Liu X, Deng W, Fu B. Identification and validation of shared gene signature of kidney renal clear cell carcinoma and COVID-19. PeerJ 2024; 12:e16927. [PMID: 38464749 PMCID: PMC10921934 DOI: 10.7717/peerj.16927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/22/2024] [Indexed: 03/12/2024] Open
Abstract
Background COVID-19 is a severe infectious disease caused by the SARS-CoV-2 virus, and previous studies have shown that patients with kidney renal clear cell carcinoma (KIRC) are more susceptible to SARS-CoV-2 infection than the general population. Nevertheless, their co-pathogenesis remains incompletely elucidated. Methods We obtained shared genes between these two diseases based on public datasets, constructed a prognostic risk model consisting of hub genes, and validated the accuracy of the model using internal and external validation sets. We further analyzed the immune landscape of the prognostic risk model, investigated the biological functions of the hub genes, and detected their expression in renal cell carcinoma cells using qPCR. Finally, we searched the candidate drugs associated with hub gene-related targets from DSigDB and CellMiner databases. Results We obtained 156 shared genes between KIRC and COVID-19 and constructed a prognostic risk model consisting of four hub genes. Both shared genes and hub genes were highly enriched in immune-related functions and pathways. Hub genes were significantly overexpressed in COVID-19 and KIRC. ROC curves, nomograms, etc., showed the reliability and robustness of the risk model, which was validated in both internal and external datasets. Moreover, patients in the high-risk group showed a higher proportion of immune cells, higher expression of immune checkpoint genes, and more active immune-related functions. Finally, we identified promising drugs for COVID-19 and KIRC, such as etoposide, fulvestrant, and topotecan. Conclusion This study identified and validated four shared genes for KIRC and COVID-19. These genes are associated with immune functions and may serve as potential prognostic biomarkers for KIRC. The shared pathways and genes may provide new insights for further mechanistic research and treatment of comorbidities.
Collapse
Affiliation(s)
- Jianqiang Nie
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hailang Yang
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqiang Liu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Wen Deng
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Bin Fu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| |
Collapse
|
8
|
Abstract
Excessive salt intake raises blood pressure, but the implications of this observation for human health have remained contentious. It has also been recognized for many years that potassium intake may mitigate the effects of salt intake on blood pressure and possibly on outcomes such as stroke. Recent large randomized intervention trials have provided strong support for the benefits of replacing salt (NaCl) with salt substitute (75% NaCl, 25% KCl) on hard outcomes, including stroke. During the same period of time, major advances have been made in understanding how the body senses and tastes salt, and how these sensations drive intake. Additionally, new insights into the complex interactions between systems that control sodium and potassium excretion by the kidneys, and the brain have highlighted the existence of a potassium switch in the kidney distal nephron. This switch seems to contribute importantly to the blood pressure-lowering effects of potassium intake. In recognition of these evolving data, the United States Food and Drug Administration is moving to permit potassium-containing salt substitutes in food manufacturing. Given that previous attempts to reduce salt consumption have not been successful, this new approach has a chance of improving health and ending the 'Salt Wars'.
Collapse
Affiliation(s)
- Robert Little
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- LeDucq Transatlantic Network of Excellence
| | - David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA
- LeDucq Transatlantic Network of Excellence
- VA Portland Health Care System, Portland, OR
| |
Collapse
|
9
|
Edwards A, Ralph DL, Mercado A, McDonough AA. Angiotensin II hypertension along the female rat tubule: predicted impact on coupled transport of Na + and K . Am J Physiol Renal Physiol 2023; 325:F733-F749. [PMID: 37823196 PMCID: PMC10878725 DOI: 10.1152/ajprenal.00232.2023] [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: 08/08/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023] Open
Abstract
Chronic infusion of subpressor level of angiotensin II (ANG II) increases the abundance of Na+ transporters along the distal nephron, balanced by suppression of Na+ transporters along the proximal tubule and medullary thick ascending limb (defined as "proximal nephron"), which impacts K+ handling along the entire renal tubule. The objective of this study was to quantitatively assess the impact of chronic ANG II on the renal handling of Na+ and K+ in female rats, using a computational model of the female rat renal tubule. Our results indicate that the downregulation of proximal nephron Na+ reabsorption (TNa), which occurs in response to ANG II-triggered hypertension, involves changes in both transporter abundance and trafficking. Our model suggests that substantial (∼30%) downregulation of active NHE3 in proximal tubule (PT) microvilli is needed to reestablish the Na+ balance at 2 wk of ANG II infusion. The 35% decrease in SGLT2, a known NHE3 regulator, may contribute to this downregulation. Both depression of proximal nephron TNa and stimulation of distal ENaC raise urinary K+ excretion in ANG II-treated females, while K+ loss is slightly mitigated by cortical NKCC2 and NCC upregulation. Our model predicts that K+ excretion may be more significantly limited during ANG II infusion by ROMK inhibition in the distal nephron and/or KCC3 upregulation in the PT, which remain open questions for experimental validation. In summary, our analysis indicates that ANG II hypertension triggers a series of events from distal TNa stimulation followed by compensatory reduction in proximal nephron TNa and accompanying adjustments to limit excessive K+ secretion.NEW & NOTEWORTHY We used a computational model of the renal tubule to assess the impact of 2-wk angiotensin II (ANG II) infusion on the handling of Na+ and K+ in female rats. ANG II strongly stimulates distal Na+ reabsorption and K+ secretion. Simulations indicate that substantial downregulation of proximal tubule NHE3 is needed to reestablish Na+ balance at 2 wk. Proximal adaptations challenge K+ homeostasis, and regulation of distal NCC and specific K+ channels likely limit urinary K+ losses.
Collapse
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Donna L Ralph
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Adriana Mercado
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| |
Collapse
|
10
|
Grimm PR, Tatomir A, Rosenbaek LL, Kim BY, Li D, Delpire EJ, Fenton RA, Welling PA. Dietary potassium stimulates Ppp1Ca-Ppp1r1a dephosphorylation of kidney NaCl cotransporter and reduces blood pressure. J Clin Invest 2023; 133:e158498. [PMID: 37676724 PMCID: PMC10617769 DOI: 10.1172/jci158498] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/06/2023] [Indexed: 09/09/2023] Open
Abstract
Consumption of low dietary potassium, common with ultraprocessed foods, activates the thiazide-sensitive sodium chloride cotransporter (NCC) via the with no (K) lysine kinase/STE20/SPS1-related proline-alanine-rich protein kinase (WNK/SPAK) pathway to induce salt retention and elevate blood pressure (BP). However, it remains unclear how high-potassium "DASH-like" diets (dietary approaches to stop hypertension) inactivate the cotransporter and whether this decreases BP. A transcriptomics screen identified Ppp1Ca, encoding PP1A, as a potassium-upregulated gene, and its negative regulator Ppp1r1a, as a potassium-suppressed gene in the kidney. PP1A directly binds to and dephosphorylates NCC when extracellular potassium is elevated. Using mice genetically engineered to constitutively activate the NCC-regulatory kinase SPAK and thereby eliminate the effects of the WNK/SPAK kinase cascade, we confirmed that PP1A dephosphorylated NCC directly in a potassium-regulated manner. Prior adaptation to a high-potassium diet was required to maximally dephosphorylate NCC and lower BP in constitutively active SPAK mice, and this was associated with potassium-dependent suppression of Ppp1r1a and dephosphorylation of its cognate protein, inhibitory subunit 1 (I1). In conclusion, potassium-dependent activation of PP1A and inhibition of I1 drove NCC dephosphorylation, providing a mechanism to explain how high dietary K+ lowers BP. Shifting signaling of PP1A in favor of activation of WNK/SPAK may provide an improved therapeutic approach for treating salt-sensitive hypertension.
Collapse
Affiliation(s)
- P. Richard Grimm
- Department of Medicine (Nephrology), Johns Hopkins University School of Medicine Baltimore, Maryland, USA
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
| | - Anamaria Tatomir
- Department of Medicine (Nephrology), Johns Hopkins University School of Medicine Baltimore, Maryland, USA
| | - Lena L. Rosenbaek
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Bo Young Kim
- Department of Medicine (Nephrology), Johns Hopkins University School of Medicine Baltimore, Maryland, USA
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
| | - Dimin Li
- Department of Medicine (Nephrology), Johns Hopkins University School of Medicine Baltimore, Maryland, USA
| | - Eric J. Delpire
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennssee, USA
| | - Robert A. Fenton
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Paul A. Welling
- Department of Medicine (Nephrology), Johns Hopkins University School of Medicine Baltimore, Maryland, USA
- The LeDucq Potassium in Hypertension Research Network of Excellence is detailed in Supplemental Acknowledgments
- Department of Physiology, Johns Hopkins University School of Medicine Baltimore, Maryland, USA
| |
Collapse
|
11
|
Kettritz R, Loffing J. Potassium homeostasis - Physiology and pharmacology in a clinical context. Pharmacol Ther 2023; 249:108489. [PMID: 37454737 DOI: 10.1016/j.pharmthera.2023.108489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Membrane voltage controls the function of excitable cells and is mainly a consequence of the ratio between the extra- and intracellular potassium concentration. Potassium homeostasis is safeguarded by balancing the extra-/intracellular distribution and systemic elimination of potassium to the dietary potassium intake. These processes adjust the plasma potassium concentration between 3.5 and 4.5 mmol/L. Several genetic and acquired diseases but also pharmacological interventions cause dyskalemias that are associated with increased morbidity and mortality. The thresholds at which serum K+ not only associates but also causes increased mortality are hotly debated. We discuss physiologic, pathophysiologic, and pharmacologic aspects of potassium regulation and provide informative case vignettes. Our aim is to help clinicians, epidemiologists, and pharmacologists to understand the complexity of the potassium homeostasis in health and disease and to initiate appropriate treatment strategies in dyskalemic patients.
Collapse
Affiliation(s)
- Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Germany.
| | | |
Collapse
|
12
|
Al-Qusairi L, Ferdaus MZ, Pham TD, Li D, Grimm PR, Zapf AM, Abood DC, Tahaei E, Delpire E, Wall SM, Welling PA. Dietary anions control potassium excretion: it is more than a poorly absorbable anion effect. Am J Physiol Renal Physiol 2023; 325:F377-F393. [PMID: 37498547 PMCID: PMC10639028 DOI: 10.1152/ajprenal.00193.2023] [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: 07/11/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 07/28/2023] Open
Abstract
The urinary potassium (K+) excretion machinery is upregulated with increasing dietary K+, but the role of accompanying dietary anions remains inadequately characterized. Poorly absorbable anions, including [Formula: see text], are thought to increase K+ secretion through a transepithelial voltage effect. Here, we tested if they also influence the K+ secretion machinery. Wild-type mice, aldosterone synthase (AS) knockout (KO) mice, or pendrin KO mice were randomized to control, high-KCl, or high-KHCO3 diets. The K+ secretory capacity was assessed in balance experiments. Protein abundance, modification, and localization of K+-secretory transporters were evaluated by Western blot analysis and confocal microscopy. Feeding the high-KHCO3 diet increased urinary K+ excretion and the transtubular K+ gradient significantly more than the high-KCl diet, coincident with more pronounced upregulation of epithelial Na+ channels (ENaC) and renal outer medullary K+ (ROMK) channels and apical localization in the distal nephron. Experiments in AS KO mice revealed that the enhanced effects of [Formula: see text] were aldosterone independent. The high-KHCO3 diet also uniquely increased the large-conductance Ca2+-activated K+ (BK) channel β4-subunit, stabilizing BKα on the apical membrane, the Cl-/[Formula: see text] exchanger, pendrin, and the apical KCl cotransporter (KCC3a), all of which are expressed specifically in pendrin-positive intercalated cells. Experiments in pendrin KO mice revealed that pendrin was required to increase K+ excretion with the high-KHCO3 diet. In summary, [Formula: see text] stimulates K+ excretion beyond a poorly absorbable anion effect, upregulating ENaC and ROMK in principal cells and BK, pendrin, and KCC3a in pendrin-positive intercalated cells. The adaptive mechanism prevents hyperkalemia and alkalosis with the consumption of alkaline ash-rich diets but may drive K+ wasting and hypokalemia in alkalosis.NEW & NOTEWORTHY Dietary anions profoundly impact K+ homeostasis. Here, we found that a K+-rich diet, containing [Formula: see text] as the counteranion, enhances the electrogenic K+ excretory machinery, epithelial Na+ channels, and renal outer medullary K+ channels, much more than a high-KCl diet. It also uniquely induces KCC3a and pendrin, in B-intercalated cells, providing an electroneutral KHCO3 secretion pathway. These findings reveal new K+ balance mechanisms that drive adaption to alkaline and K+-rich foods, which should guide new treatment strategies for K+ disorders.
Collapse
Affiliation(s)
- Lama Al-Qusairi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Mohammed Z Ferdaus
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Truyen D Pham
- Department of Medicine Nephrology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Dimin Li
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - P Richard Grimm
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ava M Zapf
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Delaney C Abood
- Department of Medicine Nephrology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Ebrahim Tahaei
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Susan M Wall
- Department of Medicine Nephrology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Zhuang L, Huang C, Ning Z, Yang L, Zou W, Wang P, Cheng CS, Meng Z. Circulating tumor-associated autoantibodies as novel diagnostic biomarkers in pancreatic adenocarcinoma. Int J Cancer 2023; 152:1013-1024. [PMID: 36274627 DOI: 10.1002/ijc.34334] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 01/06/2023]
Abstract
To develop a superior diagnostic approach for pancreatic adenocarcinoma (PAAC), the present study prospectively included 338 PAAC patients, 294 normal healthy volunteers (NHV), 122 chronic pancreatitis (CP) patients and 100 patients with non-PAAC malignancies. In the identification phase, HuProt Human Proteome Microarray, comprising 21 065 proteins, was used to identify serum tumor-associated autoantibodies (TAAbs) candidates differentiating PAAC (n = 30) from NHV (n = 30). A PAAC-focused array containing 165 differentially expressed TAAbs identified was subsequently adopted in the validation phase (n = 712) for specificity and sensitivities. The multivariate TAAbs signature for differentiation PAAC from controls (NHV + CP) identified five candidates, namely the IgG-type TAAbs against CLDN17, KCNN3, SLAMF7, SLC22A11 and OR51F2. Multivariate logistic performance model of y = (22.893 × CA19-9 + 0.68 × CLDN17 - 4.012) showed a significant better diagnostic accuracy than that of CA19-9 and CLDN17 in differentiating PAAC from controls (NHV + CP) (AUC = 0.97, 0.92 and 0.82, respectively, P-value < .0001). We further tested the autoantigen level of CLDN17 by ELISA in 82 sera samples from PAAC (n = 42), CP (n = 24) and NHV (n = 16). Similarly, the model showed superior diagnostic performance than that of CA19-9 and CLDN17 (AUC = 0.93, 0.83 and 0.81, respectively, P-value < .0001) in differentiating PAAC from controls. In conclusion, our study is the first to characterize the circulating TAAbs signatures in PAAC. The results showed that CLDN17 combined with CA19-9 provided potentially clinical value and may serve as noninvasive novel biomarkers for PAAC diagnosis.
Collapse
Affiliation(s)
- Liping Zhuang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changjing Huang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhouyu Ning
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lina Yang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Shandong Province, China
| | - Wenbin Zou
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Peng Wang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chien-Shan Cheng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Zhang C, Guo J. Diverse functions of the inward-rectifying potassium channel Kir5.1 and its relationship with human diseases. Front Physiol 2023; 14:1127893. [PMID: 36923292 PMCID: PMC10008857 DOI: 10.3389/fphys.2023.1127893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023] Open
Abstract
The inward-rectifying potassium channel subunit Kir5.1, encoded by Kcnj16, can form functional heteromeric channels (Kir4.1/5.1 and Kir4.2/5.1) with Kir4.1 (encoded by Kcnj10) or Kir4.2 (encoded by Kcnj15). It is expressed in the kidneys, pancreas, thyroid, brain, and other organs. Although Kir5.1 cannot form functional homomeric channels in most cases, an increasing number of studies in recent years have found that the functions of this subunit should not be underestimated. Kir5.1 can confer intracellular pH sensitivity to Kir4.1/5.1 channels, which can act as extracellular potassium sensors in the renal distal convoluted tubule segment. This segment plays an important role in maintaining potassium and acid-base balances. This review summarizes the various pathophysiological processes involved in Kir5.1 and the expression changes of Kir5.1 as a differentially expressed gene in various cancers, as well as describing several other disease phenotypes caused by Kir5.1 dysfunction.
Collapse
Affiliation(s)
- Chaojie Zhang
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Henan Province Research Center for Kidney Disease, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jia Guo
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Henan Province Research Center for Kidney Disease, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
17
|
Yan P, Ke B, Fang X. Ion channels as a therapeutic target for renal fibrosis. Front Physiol 2022; 13:1019028. [PMID: 36277193 PMCID: PMC9581181 DOI: 10.3389/fphys.2022.1019028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Renal ion channel transport and electrolyte disturbances play an important role in the process of functional impairment and fibrosis in the kidney. It is well known that there are limited effective drugs for the treatment of renal fibrosis, and since a large number of ion channels are involved in the renal fibrosis process, understanding the mechanisms of ion channel transport and the complex network of signaling cascades between them is essential to identify potential therapeutic approaches to slow down renal fibrosis. This review summarizes the current work of ion channels in renal fibrosis. We pay close attention to the effect of cystic fibrosis transmembrane conductance regulator (CFTR), transmembrane Member 16A (TMEM16A) and other Cl− channel mediated signaling pathways and ion concentrations on fibrosis, as well as the various complex mechanisms for the action of Ca2+ handling channels including Ca2+-release-activated Ca2+ channel (CRAC), purinergic receptor, and transient receptor potential (TRP) channels. Furthermore, we also focus on the contribution of Na+ transport such as epithelial sodium channel (ENaC), Na+, K+-ATPase, Na+-H+ exchangers, and K+ channels like Ca2+-activated K+ channels, voltage-dependent K+ channel, ATP-sensitive K+ channels on renal fibrosis. Proposed potential therapeutic approaches through further dissection of these mechanisms may provide new therapeutic opportunities to reduce the burden of chronic kidney disease.
Collapse
|
18
|
Zhu Z, Lei Z, Qian J, Zhang C, Gong Y, Yin G, Li Y, Li X, Lin J, Zhou L. The Ion Channel-Related Gene Signatures Correlated With Diagnosis, Prognosis, and Individualized Treatment in Patients With Clear Cell Renal Cell Carcinoma. Front Pharmacol 2022; 13:889142. [PMID: 35721115 PMCID: PMC9198310 DOI: 10.3389/fphar.2022.889142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Background: Early detection and precise prognostic evaluation of clear cell renal cell carcinoma (ccRCC) are crucial for patient life expectancy. Ion channel-related genes (ICRGs) are of great diagnostic and prognostic value as components that maintain the normal structure of the kidney. Therefore, we systematically explored the diagnostic, prognostic, and therapeutic value of ICRGs in ccRCC using the multi-database. Methods: RNA transcriptome profiles and clinical data of ccRCC patients were extracted and integrated from public databases including The Cancer Genome Atlas, ICGC, GEO, and E-MTAB databases. Ion channel-related genes were obtained from the literature collection. The diagnostic signature was performed using the LASSO and SVM-REF analyses. Meanwhile, the prognostic signature was conducted using the LASSO analyses. Molecular subtyping was performed using the ConsensusClusterPlus and the corresponding therapeutic targets were evaluated using the pRRophetic package. In addition, a prognostic nomogram was constructed based on the results of cox regression analyses. Results: We successfully constructed diagnostic signatures for five ICRGs and prognostic signatures for 10 ICRGs with AUC values greater than 0.7, showing good predictive performance. Based on the median risk score, we found that high-risk patients had a significantly worse prognosis. We also divided ccRCC patients into two clusters according to prognostic ICRGs, and there was a significant survival outcome between the two clusters and different sensitivity to diverse clinical therapeutic strategies. Meanwhile, we constructed a nomogram based on clinical molecules and signatures, and its predictive efficacy was better than the signature or the present tumor-node-metastasis staging system. Conclusion: In this study, we established useful signatures for early detection, prognosis evaluation, and individualized treatment for ccRCC. Moreover, KCNJ16 deserves to be explored comprehensively in the future.
Collapse
Affiliation(s)
- Zhenpeng Zhu
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhenchuan Lei
- School of Biomedical Sciences, Heart and Vascular Institute and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinqin Qian
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Cuijian Zhang
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yanqing Gong
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Guicao Yin
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yifan Li
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xuesong Li
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jian Lin
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Liqun Zhou
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
19
|
Shimamura Y, Turk M, Qader MA, Shah S, Topf JM, Hiremath S. Potassium Enriched Salt to Lower Stroke Risk: A #NephJC Editorial on the SSaSS Study. Kidney Med 2022; 4:100489. [PMID: 35801189 PMCID: PMC9253645 DOI: 10.1016/j.xkme.2022.100489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
| | - Michael Turk
- Department of Medicine, Allegheny Health Network, Pittsburgh, PA
| | - Md Abdul Qader
- Department of Pediatric Nephrology, Square Hospitals Ltd, Dhaka, Bangladesh
| | - Shweta Shah
- Renal Section, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX
| | - Joel M. Topf
- Department of Medicine, Oakland University William Beaumont School of Medicine, Auburn Hills, MI
| | - Swapnil Hiremath
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Address for Correspondence: Swapnil Hiremath, MD, MPH, Department of Medicine, University of Ottawa, 1967 Riverside Dr, Ottawa, ON K1H7W9, Canada.
| |
Collapse
|
20
|
McClenahan SJ, Kent CN, Kharade SV, Isaeva E, Williams JC, Han C, Terker A, Gresham R, Lazarenko RM, Days EL, Romaine IM, Bauer JA, Boutaud O, Sulikowski GA, Harris R, Weaver CD, Staruschenko A, Lindsley CW, Denton JS. VU6036720: The First Potent and Selective In Vitro Inhibitor of Heteromeric Kir4.1/5.1 Inward Rectifier Potassium Channels. Mol Pharmacol 2022; 101:357-370. [PMID: 35246480 PMCID: PMC9092466 DOI: 10.1124/molpharm.121.000464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/14/2022] [Indexed: 01/14/2023] Open
Abstract
Heteromeric Kir4.1/Kir5.1 (KCNJ10/KCNJ16) inward rectifier potassium (Kir) channels play key roles in the brain and kidney, but pharmacological tools for probing their physiology and therapeutic potential have not been developed. Here, we report the discovery, in a high-throughput screening of 80,475 compounds, of the moderately potent and selective inhibitor VU0493690, which we selected for characterization and chemical optimization. VU0493690 concentration-dependently inhibits Kir4.1/5.1 with an IC50 of 0.96 μM and exhibits at least 10-fold selectivity over Kir4.1 and ten other Kir channels. Multidimensional chemical optimization of VU0493690 led to the development of VU6036720, the most potent (IC50 = 0.24 μM) and selective (>40-fold over Kir4.1) Kir4.1/5.1 inhibitor reported to date. Cell-attached patch single-channel recordings revealed that VU6036720 inhibits Kir4.1/5.1 activity through a reduction of channel open-state probability and single-channel current amplitude. Elevating extracellular potassium ion by 20 mM shifted the IC50 6.8-fold, suggesting that VU6036720 is a pore blocker that binds in the ion-conduction pathway. Mutation of the "rectification controller" asparagine 161 to glutamate (N161E), which is equivalent to small-molecule binding sites in other Kir channels, led to a strong reduction of inhibition by VU6036720. Renal clearance studies in mice failed to show a diuretic response that would be consistent with inhibition of Kir4.1/5.1 in the renal tubule. Drug metabolism and pharmacokinetics profiling revealed that high VU6036720 clearance and plasma protein binding may prevent target engagement in vivo. In conclusion, VU6036720 represents the current state-of-the-art Kir4.1/5.1 inhibitor that should be useful for probing the functions of Kir4.1/5.1 in vitro and ex vivo. SIGNIFICANCE STATEMENT: Heteromeric inward rectifier potassium (Kir) channels comprising Kir4.1 and Kir5.1 subunits play important roles in renal and neural physiology and may represent inhibitory drug targets for hypertension and edema. Herein, we employ high-throughput compound library screening, patch clamp electrophysiology, and medicinal chemistry to develop and characterize the first potent and specific in vitro inhibitor of Kir4.1/5.1, VU6036720, which provides proof-of-concept that drug-like inhibitors of this channel may be developed.
Collapse
Affiliation(s)
- Samantha J McClenahan
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Caitlin N Kent
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Sujay V Kharade
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Elena Isaeva
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Jade C Williams
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Changho Han
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Andrew Terker
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Robert Gresham
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Roman M Lazarenko
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Emily L Days
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Ian M Romaine
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Joshua A Bauer
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Olivier Boutaud
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Gary A Sulikowski
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Raymond Harris
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - C David Weaver
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Alexander Staruschenko
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Craig W Lindsley
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Jerod S Denton
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| |
Collapse
|
21
|
Takei Y, Ando M, Wong MKS, Tsukada T. Molecular mechanisms underlying guanylin-induced transcellular Cl - secretion into the intestinal lumen of seawater-acclimated eels. Gen Comp Endocrinol 2022; 318:113986. [PMID: 35114197 DOI: 10.1016/j.ygcen.2022.113986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/20/2021] [Accepted: 01/27/2022] [Indexed: 11/19/2022]
Abstract
Guanylin (GN) stimulates Cl- secretion into the intestinal lumen of seawater-acclimated eels, but the molecular mechanisms of transepithelial Cl- transport are still unknown. In Ussing chamber experiments, we confirmed that mucosal application of eel GN reversed intestinal serosa-negative potential difference, indicating Cl- secretion. Serosal application of DNDS or mucosal application of DPC inhibited the GN effect, but serosal application of bumetanide had no effect. Removal of HCO3- from the serosal fluid also inhibited the GN effect. In intestinal sac experiments, mucosal GN stimulated luminal secretion of both Cl- and Na+, which was blocked by serosal DNDS. These results suggest that Cl- is taken up at the serosal side by DNDS-sensitive anion exchanger (AE) coupled with Na+-HCO3- cotransporter (NBC) but not by Na+-K+-2Cl- cotransporter 1 (NKCC1), and Cl- is secreted by unknown DPC-sensitive Cl- channel (ClC) at the mucosal side. The transcriptomic analysis combined with qPCR showed low expression of NKCC1 gene and no upregulation of the gene after seawater transfer, while high expression of ClC2 gene and upregulation after seawater transfer. In addition, SO42- transporters (apical Slc26a3/6 and basolateral Slc26a1) are also candidates for transcellular Cl- secretion in exchange of luminal SO42. Na+ secretion could occur through a paracellular route, as Na+-leaky claudin15 was highly expressed and upregulated after seawater transfer. High local Na+ concentration in the lateral interspace produced by Na+/K+-ATPase (NKA) coupled with K+ channels (Kir5.1b) seems to facilitate the paracellular transport. In situ hybridization confirmed the expression of the candidate genes in the epithelial enterocytes. Together with our previous results, we suggest that GN stimulates basolateral NBCela/AE2 and apical ClC2 to increase transcellular Cl- secretion in seawater eel intestine, which differs from the involvement of apical CFTR and basolateral NKCC1 as suggested in mammals and other teleosts.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan.
| | - Masaaki Ando
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Marty K S Wong
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Takehiro Tsukada
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
22
|
Zapf AM, Grimm PR, Al-Qusairi L, Delpire E, Welling PA. Low Salt Delivery Triggers Autocrine Release of Prostaglandin E2 From the Aldosterone-Sensitive Distal Nephron in Familial Hyperkalemic Hypertension Mice. Front Physiol 2022; 12:787323. [PMID: 35069250 PMCID: PMC8770744 DOI: 10.3389/fphys.2021.787323] [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: 09/30/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Aberrant activation of with-no-lysine kinase (WNK)-STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) kinase signaling in the distal convoluted tubule (DCT) causes unbridled activation of the thiazide-sensitive sodium chloride cotransporter (NCC), leading to familial hyperkalemic hypertension (FHHt) in humans. Studies in FHHt mice engineered to constitutively activate SPAK specifically in the DCT (CA-SPAK mice) revealed maladaptive remodeling of the aldosterone sensitive distal nephron (ASDN), characterized by decrease in the potassium excretory channel, renal outer medullary potassium (ROMK), and epithelial sodium channel (ENaC), that contributes to the hyperkalemia. The mechanisms by which NCC activation in DCT promotes remodeling of connecting tubule (CNT) are unknown, but paracrine communication and reduced salt delivery to the ASDN have been suspected. Here, we explore the involvement of prostaglandin E2 (PGE2). We found that PGE2 and the terminal PGE2 synthase, mPGES1, are increased in kidney cortex of CA-SPAK mice, compared to control or SPAK KO mice. Hydrochlorothiazide (HCTZ) reduced PGE2 to control levels, indicating increased PGE2 synthesis is dependent on increased NCC activity. Immunolocalization studies revealed mPGES1 is selectively increased in the CNT of CA-SPAK mice, implicating low salt-delivery to ASDN as the trigger. Salt titration studies in an in vitro ASDN cell model, mouse CCD cell (mCCD-CL1), confirmed PGE2 synthesis is activated by low salt, and revealed that response is paralleled by induction of mPGES1 gene expression. Finally, inhibition of the PGE2 receptor, EP1, in CA-SPAK mice partially restored potassium homeostasis as it partially rescued ROMK protein abundance, but not ENaC. Together, these data indicate low sodium delivery to the ASDN activates PGE2 synthesis and this inhibits ROMK through autocrine activation of the EP1 receptor. These findings provide new insights into the mechanism by which activation of sodium transport in the DCT causes remodeling of the ASDN.
Collapse
Affiliation(s)
- Ava M Zapf
- Molecular Medicine, Graduate Program in Life Sciences, University of Maryland Medical School, Baltimore, MD, United States
| | - Paul R Grimm
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Lama Al-Qusairi
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN, United States
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Physiology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
23
|
Pearce D, Manis AD, Nesterov V, Korbmacher C. Regulation of distal tubule sodium transport: mechanisms and roles in homeostasis and pathophysiology. Pflugers Arch 2022; 474:869-884. [PMID: 35895103 PMCID: PMC9338908 DOI: 10.1007/s00424-022-02732-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Regulated Na+ transport in the distal nephron is of fundamental importance to fluid and electrolyte homeostasis. Further upstream, Na+ is the principal driver of secondary active transport of numerous organic and inorganic solutes. In the distal nephron, Na+ continues to play a central role in controlling the body levels and concentrations of a more select group of ions, including K+, Ca++, Mg++, Cl-, and HCO3-, as well as water. Also, of paramount importance are transport mechanisms aimed at controlling the total level of Na+ itself in the body, as well as its concentrations in intracellular and extracellular compartments. Over the last several decades, the transporters involved in moving Na+ in the distal nephron, and directly or indirectly coupling its movement to that of other ions have been identified, and their interrelationships brought into focus. Just as importantly, the signaling systems and their components-kinases, ubiquitin ligases, phosphatases, transcription factors, and others-have also been identified and many of their actions elucidated. This review will touch on selected aspects of ion transport regulation, and its impact on fluid and electrolyte homeostasis. A particular focus will be on emerging evidence for site-specific regulation of the epithelial sodium channel (ENaC) and its role in both Na+ and K+ homeostasis. In this context, the critical regulatory roles of aldosterone, the mineralocorticoid receptor (MR), and the kinases SGK1 and mTORC2 will be highlighted. This includes a discussion of the newly established concept that local K+ concentrations are involved in the reciprocal regulation of Na+-Cl- cotransporter (NCC) and ENaC activity to adjust renal K+ secretion to dietary intake.
Collapse
Affiliation(s)
- David Pearce
- Department of Medicine, Division of Nephrology, and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA USA
| | - Anna D. Manis
- Department of Medicine, Division of Nephrology, and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA USA
| | - Viatcheslav Nesterov
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany, Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany, Erlangen, Germany
| |
Collapse
|
24
|
Nesterov V, Bertog M, Korbmacher C. High baseline ROMK activity in the mouse late distal convoluted and early connecting tubule probably contributes to aldosterone-independent K + secretion. Am J Physiol Renal Physiol 2022; 322:F42-F54. [PMID: 34843658 DOI: 10.1152/ajprenal.00252.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
The renal outer medullary K+ channel (ROMK) is colocalized with the epithelial Na+ channel (ENaC) in the late distal convoluted tubule (DCT2), connecting tubule (CNT), and cortical collecting duct (CCD). ENaC-mediated Na+ absorption generates the electrical driving force for ROMK-mediated tubular K+ secretion, which is critically important for maintaining renal K+ homeostasis. ENaC activity is aldosterone dependent in the late CNT and early CCD (CNT/CCD) but aldosterone independent in the DCT2 and early CNT (DCT2/CNT). This suggests that under baseline conditions with low plasma aldosterone, ROMK-mediated K+ secretion mainly occurs in the DCT2/CNT. Therefore, we hypothesized that baseline ROMK activity is higher in the DCT2/CNT than in the CNT/CCD. To test this hypothesis, patch-clamp experiments were performed in the DCT2/CNT and CNT/CCD microdissected from mice maintained on a standard diet. In single-channel recordings from outside-out patches, we detected typical ROMK channel activity in both the DCT2/CNT and CNT/CCD and confirmed that ROMK is the predominant K+ channel in the apical membrane. Amiloride-sensitive and tertiapin-sensitive whole-cell currents were determined to assess ENaC and ROMK activity, respectively. As expected, baseline amiloride-sensitive current was high in the DCT2/CNT (∼370 pA) but low in the CNT/CCD (∼60 pA). Importantly, tertiapin-sensitive current was significantly higher in the DCT2/CNT than in the CNT/CCD (∼810 vs. ∼350 pA). We conclude that high ROMK activity in the DCT2/CNT is critical for aldosterone-independent renal K+ secretion under baseline conditions. A low-K+ diet significantly reduced ENaC but not ROMK activity in the DCT2/CNT. This suggests that modifying ENaC activity in the DCT2/CNT plays a key regulatory role in adjusting renal K+ excretion to dietary K+ intake.NEW & NOTEWORTHY ROMK-mediated renal K+ secretion is essential for maintaining K+ balance and requires a lumen negative transepithelial potential critically dependent on ENaC activity. Using microdissected distal mouse tubules, we demonstrated that baseline apical ROMK activity is high in the DCT2/CNT. Aldosterone-independent baseline ENaC activity is also high in the DCT2/CNT and downregulated by a low-K+ diet, which highlights the important role of the DCT2/CNT in regulating K+ secretion in an aldosterone-independent manner.
Collapse
Affiliation(s)
- Viatcheslav Nesterov
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marko Bertog
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
25
|
Hyndman KA, Isaeva E, Palygin O, Mendoza LD, Rodan AR, Staruschenko A, Pollock JS. Role of collecting duct principal cell NOS1β in sodium and potassium homeostasis. Physiol Rep 2021; 9:e15080. [PMID: 34665521 PMCID: PMC8525323 DOI: 10.14814/phy2.15080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/15/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
The nitric oxide (NO)-generating enzyme, NO synthase-1β (NOS1β), is essential for sodium (Na+ ) homeostasis and blood pressure control. We previously showed that collecting duct principal cell NOS1β is critical for inhibition of the epithelial sodium channel (ENaC) during high Na+ intake. Previous studies on freshly isolated cortical collecting ducts (CCD) demonstrated that exogenous NO promotes basolateral potassium (K+ ) conductance through basolateral channels, presumably Kir 4.1 (Kcnj10) and Kir 5.1 (Kcnj16). We, therefore, investigated the effects of NOS1β knockout on Kir 4.1/Kir 5.1 channel activity. Indeed, in CHO cells overexpressing NOS1β and Kir 4.1/Kir 5.1, the inhibition of NO signaling decreased channel activity. Male littermate control and principal cell NOS1β knockout mice (CDNOS1KO) on a 7-day, 4% NaCl diet (HSD) were used to detect changes in basolateral K+ conductance. We previously demonstrated that CDNOS1KO mice have high circulating aldosterone despite a high-salt diet and appropriately suppressed renin. We observed greater Kir 4.1 cortical abundance and significantly greater Kir 4.1/Kir 5.1 single-channel activity in the principal cells from CDNOS1KO mice. Moreover, blocking aldosterone action with in vivo spironolactone treatment resulted in lower Kir 4.1 abundance and greater plasma K+ in the CDNOS1KO mice compared to controls. Lowering K+ content in the HSD prevented the high aldosterone and greater plasma Na+ of CDNOS1KO mice and normalized Kir 4.1 abundance. We conclude that during chronic HSD, lack of NOS1β leads to increased plasma K+ , enhanced circulating aldosterone, and activation of ENaC and Kir 4.1/Kir 5.1 channels. Thus, principal cell NOS1β is required for the regulation of both Na+ and K+ by the kidney.
Collapse
Affiliation(s)
- Kelly A. Hyndman
- Department of MedicineDivision of NephrologySection of Cardio‐Renal Physiology and MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Elena Isaeva
- Department of Cellular Biology, Neurobiology and AnatomyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Oleg Palygin
- Division of NephrologyDepartment of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Luciano D. Mendoza
- Department of MedicineDivision of NephrologySection of Cardio‐Renal Physiology and MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Aylin R. Rodan
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
- The Department of Internal MedicineDivision of Nephrology and HypertensionUniversity of UtahSalt Lake CityUtahUSA
- The Department of Human GeneticsUniversity of UtahSalt Lake CityUtahUSA
- The Medical ServiceVeterans Affairs Salt Lake City Health Care SystemSalt Lake CityUtahUSA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- The James A. Haley Veterans HospitalTampaFloridaUSA
| | - Jennifer S. Pollock
- Department of MedicineDivision of NephrologySection of Cardio‐Renal Physiology and MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
26
|
Abstract
K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).
Collapse
|
27
|
Takei Y. The digestive tract as an essential organ for water acquisition in marine teleosts: lessons from euryhaline eels. ZOOLOGICAL LETTERS 2021; 7:10. [PMID: 34154668 PMCID: PMC8215749 DOI: 10.1186/s40851-021-00175-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/16/2021] [Indexed: 05/17/2023]
Abstract
Adaptation to a hypertonic marine environment is one of the major topics in animal physiology research. Marine teleosts lose water osmotically from the gills and compensate for this loss by drinking surrounding seawater and absorbing water from the intestine. This situation is in contrast to that in mammals, which experience a net osmotic loss of water after drinking seawater. Water absorption in fishes is made possible by (1) removal of monovalent ions (desalinization) by the esophagus, (2) removal of divalent ions as carbonate (Mg/CaCO3) precipitates promoted by HCO3- secretion, and (3) facilitation of NaCl and water absorption from diluted seawater by the intestine using a suite of unique transporters. As a result, 70-85% of ingested seawater is absorbed during its passage through the digestive tract. Thus, the digestive tract is an essential organ for marine teleost survival in the hypertonic seawater environment. The eel is a species that has been frequently used for osmoregulation research in laboratories worldwide. The eel possesses many advantages as an experimental animal for osmoregulation studies, one of which is its outstanding euryhalinity, which enables researchers to examine changes in the structure and function of the digestive tract after direct transfer from freshwater to seawater. In recent years, the molecular mechanisms of ion and water transport across epithelial cells (the transcellular route) and through tight junctions (the paracellular route) have been elucidated for the esophagus and intestine. Thanks to the rapid progress in analytical methods for genome databases on teleosts, including the eel, the molecular identities of transporters, channels, pumps and junctional proteins have been clarified at the isoform level. As 10 y have passed since the previous reviews on this subject, it seems relevant and timely to summarize recent progress in research on the molecular mechanisms of water and ion transport in the digestive tract in eels and to compare the mechanisms with those of other teleosts and mammals from comparative and evolutionary viewpoints. We also propose future directions for this research field to achieve integrative understanding of the role of the digestive tract in adaptation to seawater with regard to pathways/mechanisms including the paracellular route, divalent ion absorption, metabolon formation and cellular trafficking of transporters. Notably, some of these have already attracted practical attention in laboratories.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8564, Japan.
| |
Collapse
|
28
|
Yang L, Xu Y, Gravotta D, Frindt G, Weinstein AM, Palmer LG. ENaC and ROMK channels in the connecting tubule regulate renal K+ secretion. J Gen Physiol 2021; 153:212401. [PMID: 34143184 PMCID: PMC8217949 DOI: 10.1085/jgp.202112902] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
We measured the activities of epithelial Na channels (ENaC) and ROMK channels in the distal nephron of the mouse kidney and assessed their role in the process of K+ secretion under different physiological conditions. Under basal dietary conditions (0.5% K), ENaC activity, measured as amiloride-sensitive currents, was high in cells at the distal end of the distal convoluted tubule (DCT) and proximal end of the connecting tubule (CNT), a region we call the early CNT (CNTe). In more distal parts of the CNT (aldosterone-sensitive portion [CNTas]), these currents were minimal. This functional difference correlated with alterations in the intracellular location of ENaC, which was at or near the apical membrane in CNTe and more cytoplasmic in the CNTas. ROMK activity, measured as TPNQ-sensitive currents, was substantial in both segments. A mathematical model of the rat nephron suggested that K+ secretion by the CNTe predicted from these currents provides much of the urinary K+ required for K balance on this diet. In animals fed a K-deficient diet (0.1% K), both ENaC and ROMK currents in the CNTe decreased by ∼50%, predicting a 50% decline in K+ secretion. Enhanced reabsorption by a separate mechanism is required to avoid excessive urinary K+ losses. In animals fed a diet supplemented with 3% K, ENaC currents increased modestly in the CNTe but strongly in the CNTas, while ROMK currents tripled in both segments. The enhanced secretion of K+ by the CNTe and the recruitment of secretion by the CNTas account for the additional transport required for K balance. Therefore, adaptation to increased K+ intake involves the extension of robust K+ secretion to more distal parts of the nephron.
Collapse
Affiliation(s)
- Lei Yang
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Yuanyuan Xu
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Diego Gravotta
- Department of Ophthalmology, Weill-Cornell Medical College, New York, NY
| | - Gustavo Frindt
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| | - Lawrence G Palmer
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY
| |
Collapse
|
29
|
Al-Qusairi L, Grimm PR, Zapf AM, Welling PA. Rapid development of vasopressin resistance in dietary K + deficiency. Am J Physiol Renal Physiol 2021; 320:F748-F760. [PMID: 33749322 PMCID: PMC8174811 DOI: 10.1152/ajprenal.00655.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
The association between diabetes insipidus (DI) and chronic dietary K+ deprivation is well known, but it remains uncertain how the disorder develops and whether it is influenced by the sexual dimorphism in K+ handling. Here, we determined the plasma K+ (PK) threshold for DI in male and female mice and ascertained if DI is initiated by polydipsia or by a central or nephrogenic defect. C57BL6J mice were randomized to a control diet or to graded reductions in dietary K+ for 8 days, and kidney function and transporters involved in water balance were characterized. We found that male and female mice develop polyuria and secondary polydipsia. Altered water balance coincided with a decrease in aquaporin-2 (AQP2) phosphorylation and apical localization despite increased levels of the vasopressin surrogate marker copeptin. No change in the protein abundance of urea transporter-A1 was observed. The Na+-K+-2Cl- cotransporter decreased only in males. Desmopressin treatment failed to reverse water diuresis in K+-restricted mice. These findings indicate that even a small fall in PK is associated with nephrogenic DI (NDI), coincident with the development of altered AQP2 regulation, implicating low PK as a causal trigger of NDI. We found that PK decreased more in females, and, consequently, females were more prone to develop NDI. Together, these data indicate that AQP2 regulation is disrupted by a small decrease in PK and that the response is influenced by sexual dimorphism in K+ handling. These findings provide new insights into the mechanisms linking water and K+ balances and support defining the disorder as "potassium-dependent NDI."NEW & NOTEWORTHY This study shows that aquaporin-2 regulation is disrupted by a small fall in plasma potassium levels and the response is influenced by sexual dimorphism in renal potassium handling. The findings provided new insights into the mechanisms by which water balance is altered in dietary potassium deficiency and support defining the disorder as "potassium-dependent nephrogenic diabetes insipidus."
Collapse
Affiliation(s)
- Lama Al-Qusairi
- Departments of Medicine, Nephrology, and Physiology, Johns Hopkins University Medical School, Baltimore, Maryland
| | - P Richard Grimm
- Departments of Medicine, Nephrology, and Physiology, Johns Hopkins University Medical School, Baltimore, Maryland
| | - Ava M Zapf
- Graduate Program in Life Sciences, University of Maryland, Baltimore, Maryland
| | - Paul A Welling
- Departments of Medicine, Nephrology, and Physiology, Johns Hopkins University Medical School, Baltimore, Maryland
| |
Collapse
|
30
|
Mukherjee A, Yang CL, McCormick JA, Martz K, Sharma A, Ellison DH. Roles of WNK4 and SPAK in K +-mediated dephosphorylation of the NaCl cotransporter. Am J Physiol Renal Physiol 2021; 320:F719-F733. [PMID: 33719576 PMCID: PMC8174808 DOI: 10.1152/ajprenal.00459.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Phosphorylation of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) is altered rapidly in response to changes in extracellular K+ concentration ([K+]). High extracellular [K+] is believed to activate specific phosphatases to dephosphorylate NCC, thereby reducing its activity. This process is defective in the human disease familial hyperkalemic hypertension, in which extracellular [K+] fails to dephosphorylate NCC, suggesting an interplay between NCC-activating and NCC-inactivating switches. Here, we explored the role of STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) and intracellular Cl- concentration in the rapid effects of extracellular K+ on NCC phosphorylation. SPAK was found to be rapidly dephosphorylated in vitro in human embryonic kidney cells and ex vivo in kidney slices by high [K+]. Acute high-K+ challenge resulted in DCT1-specific SPAK dephosphorylation in vivo and dissolution of SPAK puncta. In line with the postulate of interplay between activating and inactivating switches, we found that the "on" switch, represented by with no lysine kinase 4 (WNK4)-SPAK, must be turned off for rapid NCC dephosphorylation by high [K+]. Longer-term WNK-SPAK-mediated stimulation, however, altered the sensitivity of the system, as it attenuated rapid NCC dephosphorylation due to acute K+ loading. Although blockade of protein phosphatase (PP)1 increased NCC phosphorylation at baseline, neither PP1 nor PP3, singly or in combination, was essential for NCC dephosphorylation. Overall, our data suggest that NCC phosphorylation is regulated by a dynamic equilibrium between activating kinases and inactivating phosphatases, with kinase inactivation playing a key role in the rapid NCC dephosphorylation by high extracellular K+.NEW & NOTEWORTHY Although a great deal is known about mechanisms by which thiazide-sensitive NaCl cotransporter is phosphorylated and activated, much less is known about dephosphorylation. Here, we show that rapid dephosphorylation by high K+ depends on the Cl- sensitivity of with no lysine kinase 4 and the rapid dephosphorylation of STE20/SPS1-related proline-alanine-rich protein kinase, primarily along the early distal convoluted tubule.
Collapse
Affiliation(s)
- Anindit Mukherjee
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Kevin Martz
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon
- Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
31
|
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 2021; 8:272-278. [PMID: 33997174 PMCID: PMC8093645 DOI: 10.1016/j.gendis.2020.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [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
|
32
|
Tamargo J, Caballero R, Delpón E. The pharmacotherapeutic management of hyperkalemia in patients with cardiovascular disease. Expert Opin Pharmacother 2021; 22:1319-1341. [PMID: 33620275 DOI: 10.1080/14656566.2021.1891223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Patients with cardiovascular diseases (CVD) are at increased risk of hyperkalemia, particularly when treated with renin-angiotensin-aldosterone inhibitors (RAASIs). Because the occurrence or fear of hyperkalemia, RAASIs are frequently down-titrated or discontinued in patients with CVD, with consequent worse outcomes than patients who remain on maximum doses.Areas covered: This article reviews potassium homeostasis, epidemiology, risk factors, and outcomes of hyperkalemia, and efficacy and safety of the drugs used for acute and chronic treatment of hyperkalemia. A literature search was carried out using the PubMed and guidelines for the management of hyperkalemia.Expert opinion: The emergency treatment of hyperkalemia is not supported by high-quality evidence and clinical trials did not report drug effects on clinical outcomes. Two potassium binders, patiromer and sodium zirconium cyclosilicate, represent a new approach in the treatment of chronic hyperkalemia as they may allow the titration and maintenance of guidelines-recommended doses of RAASIs in patients with CVD who otherwise would not tolerate them due to the risk of hyperkalemia.Further studies are needed to evaluate the safety and efficacy of drug therapy and support the development of guidelines for acute and chronic hyperkalemia.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Instituto De Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Instituto De Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Instituto De Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
33
|
Weaver CD, Denton JS. Next-generation inward rectifier potassium channel modulators: discovery and molecular pharmacology. Am J Physiol Cell Physiol 2021; 320:C1125-C1140. [PMID: 33826405 DOI: 10.1152/ajpcell.00548.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inward rectifying potassium (Kir) channels play important roles in both excitable and nonexcitable cells of various organ systems and could represent valuable new drug targets for cardiovascular, metabolic, immune, and neurological diseases. In nonexcitable epithelial cells of the kidney tubule, for example, Kir1.1 (KCNJ1) and Kir4.1 (KCNJ10) are linked to sodium reabsorption in the thick ascending limb of Henle's loop and distal convoluted tubule, respectively, and have been explored as novel-mechanism diuretic targets for managing hypertension and edema. G protein-coupled Kir channels (Kir3) channels expressed in the central nervous system are critical effectors of numerous signal transduction pathways underlying analgesia, addiction, and respiratory-depressive effects of opioids. The historical dearth of pharmacological tool compounds for exploring the therapeutic potential of Kir channels has led to a molecular target-based approach using high-throughput screen (HTS) of small-molecule libraries and medicinal chemistry to develop "next-generation" Kir channel modulators that are both potent and specific for their targets. In this article, we review recent efforts focused specifically on discovery and improvement of target-selective molecular probes. The reader is introduced to fluorescence-based thallium flux assays that have enabled much of this work and then provided with an overview of progress made toward developing modulators of Kir1.1 (VU590, VU591), Kir2.x (ML133), Kir3.X (ML297, GAT1508, GiGA1, VU059331), Kir4.1 (VU0134992), and Kir7.1 (ML418). We discuss what is known about the small molecules' molecular mechanisms of action, in vitro and in vivo pharmacology, and then close with our view of what critical work remains to be done.
Collapse
Affiliation(s)
- C David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Jerod S Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
34
|
Chen L, Chou CL, Knepper MA. Targeted Single-Cell RNA-seq Identifies Minority Cell Types of Kidney Distal Nephron. J Am Soc Nephrol 2021; 32:886-896. [PMID: 33769948 PMCID: PMC8017539 DOI: 10.1681/asn.2020101407] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/03/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Proximal tubule cells dominate the kidney parenchyma numerically, although less abundant cell types of the distal nephron have disproportionate roles in water and electrolyte balance. METHODS Coupling of a FACS-based enrichment protocol with single-cell RNA-seq profiled the transcriptomes of 9099 cells from the thick ascending limb (CTAL)/distal convoluted tubule (DCT) region of the mouse nephron. RESULTS Unsupervised clustering revealed Slc12a3 +/Pvalb + and Slc12a3 +/Pvalb - cells, identified as DCT1 and DCT2 cells, respectively. DCT1 cells appear to be heterogeneous, with orthogonally variable expression of Slc8a1, Calb1, and Ckb. An additional DCT1 subcluster showed marked enrichment of cell cycle-/cell proliferation-associated mRNAs (e.g., Mki67, Stmn1, and Top2a), which fit with the known plasticity of DCT cells. No DCT2-specific transcripts were found. DCT2 cells contrast with DCT1 cells by expression of epithelial sodium channel β- and γ-subunits and much stronger expression of transcripts associated with calcium transport (Trpv5, Calb1, S100g, and Slc8a1). Additionally, scRNA-seq identified three distinct CTAL (Slc12a1 +) cell subtypes. One of these expressed Nos1 and Avpr1a, consistent with macula densa cells. The other two CTAL clusters were distinguished by Cldn10 and Ptger3 in one and Cldn16 and Foxq1 in the other. These two CTAL cell types were also distinguished by expression of alternative Iroquois homeobox transcription factors, with Irx1 and Irx2 in the Cldn10 + CTAL cells and Irx3 in the Cldn16 + CTAL cells. CONCLUSIONS Single-cell transcriptomics revealed unexpected diversity among the cells of the distal nephron in mouse. Web-based data resources are provided for the single-cell data.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
35
|
Chen L, Chou CL, Knepper MA. A Comprehensive Map of mRNAs and Their Isoforms across All 14 Renal Tubule Segments of Mouse. J Am Soc Nephrol 2021; 32:897-912. [PMID: 33769951 PMCID: PMC8017530 DOI: 10.1681/asn.2020101406] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/13/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The repertoire of protein expression along the renal tubule depends both on regulation of transcription and regulation of alternative splicing that can generate multiple proteins from a single gene. METHODS A full-length, small-sample RNA-seq protocol profiled transcriptomes for all 14 renal tubule segments microdissected from mouse kidneys. RESULTS This study identified >34,000 transcripts, including 3709 that were expressed in a segment-specific manner. All data are provided as an online resource (https://esbl.nhlbi.nih.gov/MRECA/Nephron/). Many of the genes expressed in unique patterns along the renal tubule were solute carriers, transcription factors, or G protein-coupled receptors that account for segment-specific function. Mapping the distribution of transcripts associated with Wnk-SPAK-PKA signaling, renin-angiotensin-aldosterone signaling, and cystic diseases of the kidney illustrated the applications of the online resource. The method allowed full-length mapping of RNA-seq reads, which facilitated comprehensive, unbiased characterization of alternative exon usage along the renal tubule, including known isoforms of Cldn10, Kcnj1 (ROMK), Slc12a1 (NKCC2), Wnk1, Stk39 (SPAK), and Slc14a2 (UT-A urea transporter). It also identified many novel isoforms with segment-specific distribution. These included variants associated with altered protein structure (Slc9a8, Khk, Tsc22d1, and Scoc), and variants that may affect untranslated, regulatory regions of transcripts (Pth1r, Pkar1a, and Dab2). CONCLUSIONS Full-length, unbiased sequencing of transcripts identified gene-expression patterns along the mouse renal tubule. The data, provided as an online resource, include both quantitative and qualitative differences in transcripts. Identification of alternative splicing along the renal tubule may prove critical to understanding renal physiology and pathophysiology.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
36
|
Murillo-de-Ozores AR, Rodríguez-Gama A, Carbajal-Contreras H, Gamba G, Castañeda-Bueno M. WNK4 kinase: from structure to physiology. Am J Physiol Renal Physiol 2021; 320:F378-F403. [PMID: 33491560 DOI: 10.1152/ajprenal.00634.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With no lysine kinase-4 (WNK4) belongs to a serine-threonine kinase family characterized by the atypical positioning of its catalytic lysine. Despite the fact that WNK4 has been found in many tissues, the majority of its study has revolved around its function in the kidney, specifically as a positive regulator of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule of the nephron. This is explained by the description of gain-of-function mutations in the gene encoding WNK4 that causes familial hyperkalemic hypertension. This disease is mainly driven by increased downstream activation of the Ste20/SPS1-related proline-alanine-rich kinase/oxidative stress responsive kinase-1-NCC pathway, which increases salt reabsorption in the distal convoluted tubule and indirectly impairs renal K+ secretion. Here, we review the large volume of information that has accumulated about different aspects of WNK4 function. We first review the knowledge on WNK4 structure and enumerate the functional domains and motifs that have been characterized. Then, we discuss WNK4 physiological functions based on the information obtained from in vitro studies and from a diverse set of genetically modified mouse models with altered WNK4 function. We then review in vitro and in vivo evidence on the different levels of regulation of WNK4. Finally, we go through the evidence that has suggested how different physiological conditions act through WNK4 to modulate NCC activity.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | | | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| |
Collapse
|
37
|
Ayasse N, Berg P, Leipziger J, Sørensen MV. ENaC expression correlates with the acute furosemide-induced K + excretion. Physiol Rep 2021; 9:e14668. [PMID: 33410279 PMCID: PMC7788322 DOI: 10.14814/phy2.14668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In the aldosterone-sensitive distal nephron (ASDN), epithelial sodium channel (ENaC)-mediated Na+ absorption drives K+ excretion. K+ excretion depends on the delivery of Na+ to the ASDN and molecularly activated ENaC. Furosemide is known as a K+ wasting diuretic as it greatly enhances Na+ delivery to the ASDN. Here, we studied the magnitude of acute furosemide-induced kaliuresis under various states of basal molecular ENaC activity. METHODS C57/Bl6J mice were subjected to different dietary regimens that regulate molecular ENaC expression and activity levels. The animals were anesthetized and bladder-catheterized. Diuresis was continuously measured before and after administration of furosemide (2 µg/g BW) or benzamil (0.2 µg/g BW). Flame photometry was used to measure urinary [Na+ ] and [K+ ]. The kidneys were harvested and, subsequently, ENaC expression and cleavage activation were determined by semiquantitative western blotting. RESULTS A low K+ and a high Na+ diet markedly suppressed ENaC protein expression, cleavage activation, and furosemide-induced kaliuresis. In contrast, furosemide-induced kaliuresis was greatly enhanced in animals fed a high K+ or low Na+ diet, conditions with increased ENaC expression. The furosemide-induced diuresis was similar in all dietary groups. CONCLUSION Acute furosemide-induced kaliuresis differs greatly and depends on the a priori molecular expression level of ENaC. Remarkably, it can be even absent in animals fed a high Na+ diet, despite a marked increase of tubular flow and urinary Na+ excretion. This study provides auxiliary evidence that acute ENaC-dependent K+ excretion requires both Na+ as substrate and molecular activation of ENaC.
Collapse
Affiliation(s)
- Niklas Ayasse
- Department of Biomedicine, PhysiologyAarhus UniversityAarhus CDenmark
| | - Peder Berg
- Department of Biomedicine, PhysiologyAarhus UniversityAarhus CDenmark
| | - Jens Leipziger
- Department of Biomedicine, PhysiologyAarhus UniversityAarhus CDenmark
- Aarhus Institute of Advanced StudiesAarhus UniversityAarhus CDenmark
| | | |
Collapse
|
38
|
Okada M, Kozaki I, Honda H. Antidepressive effect of an inward rectifier K+ channel blocker peptide, tertiapin-RQ. PLoS One 2020; 15:e0233815. [PMID: 33186384 PMCID: PMC7665585 DOI: 10.1371/journal.pone.0233815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/13/2020] [Indexed: 11/18/2022] Open
Abstract
Renal outer medullary K+ channel, ROMK (Kir1.1, kcnj1) is expressed in the kidney and brain, but its role in the central nervous system remains unknown. Recent studies suggested an involvement of the ROMK channel in mental diseases. Tertiapin (TPN) is a European honey bee venom peptide and is reported to selectively block the ROMK channel. Here, we have chemically synthesized a series of mutated TPN peptides, including TPN-I8R and -M13Q (TPN-RQ), reported previously, and examined their blocking activity on the ROMK channel. Among 71 peptides tested, TPN-RQ was found to block the ROMK channel most effectively. Whole-cell patch-clamp recordings showed the essential roles of two disulfide bonds and the circular structure for the blockade activity. To examine the central role, we injected TPN-RQ intracerebroventricularly and examined the effects on depression- and anxiety-like behaviors in mice. TPN-RQ showed an antidepressive effect in tail-suspension and forced swim tests. The injection of TPN-RQ also enhanced the anxiety-like behavior in the elevated plus-maze and light/dark box tests and impaired spontaneous motor activities in balance beam and wheel running tests. Administration of TPM-RQ suppressed the anti-c-Fos immunoreactivity in the lateral septum, without affecting immunoreactivity in antidepressant-related nuclei, e.g. the dorsal raphe nucleus and locus coeruleus. TPN-RQ may exert its antidepressive effects through a different mechanism from current drugs.
Collapse
Affiliation(s)
- Masayoshi Okada
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, Kurashiki, Okayama, Japan
- * E-mail:
| | - Ikkou Kozaki
- Department of Biomolecular Engineering, Graduate Schoosl of Engineering, Nagoya University, Nagoya, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate Schoosl of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
39
|
Song A, Wang J, Tong Y, Fang J, Zhang Y, Zhang H, Ruan H, Wang K, Liu Y. BKCa channels regulate the immunomodulatory properties of WJ-MSCs by affecting the exosome protein profiles during the inflammatory response. Stem Cell Res Ther 2020; 11:440. [PMID: 33059770 PMCID: PMC7560248 DOI: 10.1186/s13287-020-01952-9] [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: 05/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) from the human umbilical cord have been studied extensively due to their immunomodulatory functions. Large-conductance Ca2+-activated K+ (BKCa channels) channels are involved in many inflammatory responses, but their involvement in the anti-inflammatory activity of WJ-MSCs is unknown. The underlying molecular mechanism, through which BKCa channels mediate the immunomodulation of WJ-MSC, which may include changes in exosomes proteomics, has not yet been clarified. Methods Alizarin staining, Oil Red O staining, and flow cytometry were used to identify WJ-MSCs, which were isolated from human umbilical cord Wharton’s jelly. BKCa channels were detected in WJ-MSCs using western blotting, real-time polymerase chain reaction (real-time PCR), and electrophysiology, and cytokine expression was examined using real-time PCR and enzyme-linked immunosorbent assays (ELISAs). Exosomes were characterized using transmission electron microscopy and nanoparticle tracking analysis. Proteomics analysis was performed to explore exosomal proteomic profiles. Results The cells derived from human umbilical cord Wharton’s jelly were identified as MSCs. BKCa channels were detected in the isolated WJ-MSCs, and the expression of these channels increased after lipopolysaccharide (LPS) stimulation. BKCa channels blockade in LPS-treated WJ-MSCs induced apoptosis and decreased interleukin-6 (IL-6) expression. Furthermore, THP-1 cells (human monocytic cell line) stimulated with LPS/interferon gamma (IFN-γ) produced more anti-inflammatory cytokines after treatment with exosomes derived from BKCa channel-knockdown WJ-MSCs (si-exo). We also observed altered expression of mitochondrial ATP synthase alpha subunit (ATP5A1), filamin B, and other proteins in si-exo, which might increase the anti-inflammatory activity of macrophages. Conclusions Our study described the functional expression of BKCa channels in WJ-MSCs, and BKCa channels regulated the immunomodulatory properties of WJ-MSCs by affecting the exosomal protein profiles during the inflammatory response.
Collapse
Affiliation(s)
- Ahui Song
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Jingjing Wang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yan Tong
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Junyan Fang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yi Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Huiping Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Hongqiang Ruan
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Kai Wang
- The Clinical and Translational Research Center Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yingli Liu
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China.
| |
Collapse
|
40
|
Esposito P, Conti NE, Falqui V, Cipriani L, Picciotto D, Costigliolo F, Garibotto G, Saio M, Viazzi F. New Treatment Options for Hyperkalemia in Patients with Chronic Kidney Disease. J Clin Med 2020; 9:2337. [PMID: 32707890 PMCID: PMC7465118 DOI: 10.3390/jcm9082337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/09/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Hyperkalemia may cause life-threatening cardiac and neuromuscular alterations, and it is associated with high mortality rates. Its treatment includes a multifaceted approach, guided by potassium levels and clinical presentation. In general, treatment of hyperkalemia may be directed towards stabilizing cell membrane potential, promoting transcellular potassium shift and lowering total K+ body content. The latter can be obtained by dialysis, or by increasing potassium elimination by urine or the gastrointestinal tract. Until recently, the only therapeutic option for increasing fecal K+ excretion was represented by the cation-exchanging resin sodium polystyrene sulfonate. However, despite its common use, the efficacy of this drug has been poorly studied in controlled studies, and concerns about its safety have been reported. Interestingly, new drugs, namely patiromer and sodium zirconium cyclosilicate, have been developed to treat hyperkalemia by increasing gastrointestinal potassium elimination. These medications have proved their efficacy and safety in large clinical trials, involving subjects at high risk of hyperkalemia, such as patients with heart failure and chronic kidney disease. In this review, we discuss the mechanisms of action and the updated data of patiromer and sodium zirconium cyclosilicate, considering that the availability of these new treatment options offers the possibility of improving the management of both acute and chronic hyperkalemia.
Collapse
Affiliation(s)
- Pasquale Esposito
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Viale Benedetto XV, 16132 Genoa, Italy; (N.E.C.); (V.F.); (L.C.); (D.P.); (F.C.); (G.G.); (M.S.); (F.V.)
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bi Y, Li C, Zhang Y, Wang Y, Chen S, Yue Q, Hoover RS, Wang XH, Delpire E, Eaton DC, Zhuang J, Cai H. Stimulatory Role of SPAK Signaling in the Regulation of Large Conductance Ca 2+-Activated Potassium (BK) Channel Protein Expression in Kidney. Front Physiol 2020; 11:638. [PMID: 32714200 PMCID: PMC7343913 DOI: 10.3389/fphys.2020.00638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
SPS1-related proline/alanine-rich kinase (SPAK) plays important roles in regulating the function of numerous ion channels and transporters. With-no-lysine (WNK) kinase phosphorylates SPAK kinase to active the SPAK signaling pathway. Our previous studies indicated that WNK kinases regulate the activity of the large-conductance Ca2+-activated K+ (BK) channel and its protein expression via the ERK1/2 signaling pathway. It remains largely unknown whether SPAK kinase directly modulates the BK protein expression in kidney. In this study, we investigated the effect of SPAK on renal BK protein expression in both HEK293 cells and mouse kidney. In HEK293 cells, siRNA-mediated knockdown of SPAK expression significantly reduced BK protein expression and increased ERK1/2 phosphorylation, whereas overexpression of SPAK significantly enhanced BK expression and decreased ERK1/2 phosphorylation in a dose-dependent manner. Knockdown of ERK1/2 prevented SPAK siRNA-mediated inhibition of BK expression. Similarly, pretreatment of HEK293 cells with either the lysosomal inhibitor bafilomycin A1 or the proteasomal inhibitor MG132 reversed the inhibitory effects of SPAK knockdown on BK expression. We also found that there is no BK channel activity in PCs of CCD in SPAK KO mice using the isolated split-open tubule single-cell patching. In addition, we found that BK protein abundance in the kidney of SPAK knockout mice was significantly decreased and ERK1/2 phosphorylation was significantly enhanced. A high-potassium diet significantly increased BK protein abundance and SPAK phosphorylation levels, while reducing ERK1/2 phosphorylation levels. These findings suggest that SPAK enhances BK protein expression by reducing ERK1/2 signaling-mediated lysosomal and proteasomal degradations of the BK channel.
Collapse
Affiliation(s)
- Ye Bi
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China.,Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Chunmei Li
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Yiqian Zhang
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunman Wang
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Shan Chen
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Qiang Yue
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Robert S Hoover
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN, United States
| | - Douglas C Eaton
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jieqiu Zhuang
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
| |
Collapse
|
42
|
Mabillard H, Sayer JA. SARS-CoV-2 and hypokalaemia: evidence and implications. F1000Res 2020; 9:587. [PMID: 33093945 PMCID: PMC7533733 DOI: 10.12688/f1000research.24441.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 01/11/2024] Open
Abstract
The global pandemic secondary to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is leading to unprecedented global morbidity and mortality. With a bewildering array of complications, renal involvement in various forms is common, including serum electrolyte derangements. Hypokalaemia secondary to SARS-CoV-2 was common in a reported Chinese cohort. Here we review the emerging evidence on hypokalaemia and SARS-CoV-2 infection, the potential pathophysiological mechanisms based on early clinical and histopathological data and important clinical implications. Mechanisms of hypokalaemia are multifactorial and so the electrolyte disturbance can be difficult to avoid. We provide further support to the theory of renin-angiotensin-aldosterone (RAS) activation, discuss the strengths and weaknesses of implicating RAS involvement and highlight the importance of calculating the transtubular potassium gradient to identify those at risk of hypokalaemia and its complications.
Collapse
Affiliation(s)
- Holly Mabillard
- Renal Services, The Newcastle Hospitals NHS Foundation Trust, Newacstle upon Tyne, Tyne and Wear, NE77DN, UK
| | - John A. Sayer
- Renal Services, The Newcastle Hospitals NHS Foundation Trust, Newacstle upon Tyne, Tyne and Wear, NE77DN, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, NE13BZ, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, Tyne and Wear, NE45PL, UK
| |
Collapse
|
43
|
Abstract
The global pandemic secondary to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is leading to unprecedented global morbidity and mortality. With a bewildering array of complications, renal involvement in various forms is common, including serum electrolyte derangements. Hypokalaemia secondary to SARS-CoV-2 was common in a reported Chinese cohort. Here we review the emerging evidence on hypokalaemia and SARS-CoV-2 infection, the potential pathophysiological mechanisms based on early clinical and histopathological data and important clinical implications. Mechanisms of hypokalaemia are multifactorial and so the electrolyte disturbance can be difficult to avoid. We provide further support to the theory of renin-angiotensin-aldosterone (RAS) activation, discuss the strengths and weaknesses of implicating RAS involvement and highlight the importance of calculating the transtubular potassium gradient to identify those at risk of hypokalaemia and its complications.
Collapse
Affiliation(s)
- Holly Mabillard
- Renal Services, The Newcastle Hospitals NHS Foundation Trust, Newacstle upon Tyne, Tyne and Wear, NE77DN, UK
| | - John A. Sayer
- Renal Services, The Newcastle Hospitals NHS Foundation Trust, Newacstle upon Tyne, Tyne and Wear, NE77DN, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, NE13BZ, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, Tyne and Wear, NE45PL, UK
| |
Collapse
|
44
|
Abstract
Ca2+- and voltage-gated K+ channels of large conductance (BK channels) are expressed in a diverse variety of both excitable and inexcitable cells, with functional properties presumably uniquely calibrated for the cells in which they are found. Although some diversity in BK channel function, localization, and regulation apparently arises from cell-specific alternative splice variants of the single pore-forming α subunit ( KCa1.1, Kcnma1, Slo1) gene, two families of regulatory subunits, β and γ, define BK channels that span a diverse range of functional properties. We are just beginning to unravel the cell-specific, physiological roles served by BK channels of different subunit composition.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
45
|
Hunter RW, Bailey MA. Hyperkalemia: pathophysiology, risk factors and consequences. Nephrol Dial Transplant 2020; 34:iii2-iii11. [PMID: 31800080 PMCID: PMC6892421 DOI: 10.1093/ndt/gfz206] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
There have been significant recent advances in our understanding of the mechanisms that maintain potassium homoeostasis and the clinical consequences of hyperkalemia. In this article we discuss these advances within a concise review of the pathophysiology, risk factors and consequences of hyperkalemia. We highlight aspects that are of particular relevance for clinical practice. Hyperkalemia occurs when renal potassium excretion is limited by reductions in glomerular filtration rate, tubular flow, distal sodium delivery or the expression of aldosterone-sensitive ion transporters in the distal nephron. Accordingly, the major risk factors for hyperkalemia are renal failure, diabetes mellitus, adrenal disease and the use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers or potassium-sparing diuretics. Hyperkalemia is associated with an increased risk of death, and this is only in part explicable by hyperkalemia-induced cardiac arrhythmia. In addition to its well-established effects on cardiac excitability, hyperkalemia could also contribute to peripheral neuropathy and cause renal tubular acidosis. Hyperkalemia-or the fear of hyperkalemia-contributes to the underprescription of potentially beneficial medications, particularly in heart failure. The newer potassium binders could play a role in attempts to minimize reduced prescribing of renin-angiotensin inhibitors and mineraolocorticoid antagonists in this context.
Collapse
Affiliation(s)
- Robert W Hunter
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK
| | - Matthew A Bailey
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK
| |
Collapse
|
46
|
Walsh KB. Screening Technologies for Inward Rectifier Potassium Channels: Discovery of New Blockers and Activators. SLAS DISCOVERY 2020; 25:420-433. [PMID: 32292089 DOI: 10.1177/2472555220905558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
K+ channels play a critical role in maintaining the normal electrical activity of excitable cells by setting the cell resting membrane potential and by determining the shape and duration of the action potential. In nonexcitable cells, K+ channels establish electrochemical gradients necessary for maintaining salt and volume homeostasis of body fluids. Inward rectifier K+ (Kir) channels typically conduct larger inward currents than outward currents, resulting in an inwardly rectifying current versus voltage relationship. This property of inward rectification results from the voltage-dependent block of the channels by intracellular polyvalent cations and makes these channels uniquely designed for maintaining the resting potential near the K+ equilibrium potential (EK). The Kir family of channels consist of seven subfamilies of channels (Kir1.x through Kir7.x) that include the classic inward rectifier (Kir2.x) channel, the G-protein-gated inward rectifier K+ (GIRK) (Kir3.x), and the adenosine triphosphate (ATP)-sensitive (KATP) (Kir 6.x) channels as well as the renal Kir1.1 (ROMK), Kir4.1, and Kir7.1 channels. These channels not only function to regulate electrical/electrolyte transport activity, but also serve as effector molecules for G-protein-coupled receptors (GPCRs) and as molecular sensors for cell metabolism. Of significance, Kir channels represent promising pharmacological targets for treating a number of clinical conditions, including cardiac arrhythmias, anxiety, chronic pain, and hypertension. This review provides a brief background on the structure, function, and pharmacology of Kir channels and then focuses on describing and evaluating current high-throughput screening (HTS) technologies, such as membrane potential-sensitive fluorescent dye assays, ion flux measurements, and automated patch clamp systems used for Kir channel drug discovery.
Collapse
Affiliation(s)
- Kenneth B Walsh
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
47
|
Ponzoni L, Nguyen NH, Bahar I, Brodsky JL. Complementary computational and experimental evaluation of missense variants in the ROMK potassium channel. PLoS Comput Biol 2020; 16:e1007749. [PMID: 32251469 PMCID: PMC7162551 DOI: 10.1371/journal.pcbi.1007749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 04/16/2020] [Accepted: 02/25/2020] [Indexed: 02/02/2023] Open
Abstract
The renal outer medullary potassium (ROMK) channel is essential for potassium transport in the kidney, and its dysfunction is associated with a salt-wasting disorder known as Bartter syndrome. Despite its physiological significance, we lack a mechanistic understanding of the molecular defects in ROMK underlying most Bartter syndrome-associated mutations. To this end, we employed a ROMK-dependent yeast growth assay and tested single amino acid variants selected by a series of computational tools representative of different approaches to predict each variants’ pathogenicity. In one approach, we used in silico saturation mutagenesis, i.e. the scanning of all possible single amino acid substitutions at all sequence positions to estimate their impact on function, and then employed a new machine learning classifier known as Rhapsody. We also used two additional tools, EVmutation and Polyphen-2, which permitted us to make consensus predictions on the pathogenicity of single amino acid variants in ROMK. Experimental tests performed for selected mutants in different classes validated the vast majority of our predictions and provided insights into variants implicated in ROMK dysfunction. On a broader scope, our analysis suggests that consolidation of data from complementary computational approaches provides an improved and facile method to predict the severity of an amino acid substitution and may help accelerate the identification of disease-causing mutations in any protein. As the number of sequenced human genomes rises, a major challenge is to identify which single amino acid variations in a protein affect function and predispose individuals to disease. While predictive algorithms are available for this purpose, a comparative analysis of recently developed algorithms has not been adequately performed, nor is it clear whether combining algorithms would improve predictive power. To this end, we compared the efficacy of three publicly available algorithms and applied the results to Bartter syndrome, a human disease for which numerous poorly-characterized single amino acid variants have been identified and for which there is no cure. In silico saturation mutagenesis, i.e., the computational prediction of pathogenesis for every possible amino acid substitution, allowed us to experimentally test predictions by measuring the activity of an ion channel linked to Bartter syndrome. Based on data from blinded experiments, we discovered that Rhapsody and EVmutation successfully predicted deleterious mutations. Moreover, Rhapsody—which takes into account evolutionary as well as structural and dynamic considerations—predicted that >90% of known Bartter syndrome mutations are deleterious. Overall, our data will aid investigators who wish to test single amino acid variants in any protein and aid biomedical researchers who wish to develop hypotheses on the potential severity of genetic variants uncovered from genome databases.
Collapse
Affiliation(s)
- Luca Ponzoni
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nga H. Nguyen
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (IB); (JLB)
| | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (IB); (JLB)
| |
Collapse
|
48
|
Recent insights into sodium and potassium handling by the aldosterone-sensitive distal nephron: implications on pathophysiology and drug discovery. J Nephrol 2020; 33:447-466. [DOI: 10.1007/s40620-020-00700-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 01/02/2020] [Indexed: 12/31/2022]
|
49
|
Recent insights into sodium and potassium handling by the aldosterone-sensitive distal nephron: a review of the relevant physiology. J Nephrol 2020; 33:431-445. [DOI: 10.1007/s40620-019-00684-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
|
50
|
Hoorn EJ, Gritter M, Cuevas CA, Fenton RA. Regulation of the Renal NaCl Cotransporter and Its Role in Potassium Homeostasis. Physiol Rev 2020; 100:321-356. [PMID: 31793845 DOI: 10.1152/physrev.00044.2018] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Daily dietary potassium (K+) intake may be as large as the extracellular K+ pool. To avoid acute hyperkalemia, rapid removal of K+ from the extracellular space is essential. This is achieved by translocating K+ into cells and increasing urinary K+ excretion. Emerging data now indicate that the renal thiazide-sensitive NaCl cotransporter (NCC) is critically involved in this homeostatic kaliuretic response. This suggests that the early distal convoluted tubule (DCT) is a K+ sensor that can modify sodium (Na+) delivery to downstream segments to promote or limit K+ secretion. K+ sensing is mediated by the basolateral K+ channels Kir4.1/5.1, a capacity that the DCT likely shares with other nephron segments. Thus, next to K+-induced aldosterone secretion, K+ sensing by renal epithelial cells represents a second feedback mechanism to control K+ balance. NCC’s role in K+ homeostasis has both physiological and pathophysiological implications. During hypovolemia, NCC activation by the renin-angiotensin system stimulates Na+ reabsorption while preventing K+ secretion. Conversely, NCC inactivation by high dietary K+ intake maximizes kaliuresis and limits Na+ retention, despite high aldosterone levels. NCC activation by a low-K+ diet contributes to salt-sensitive hypertension. K+-induced natriuresis through NCC offers a novel explanation for the antihypertensive effects of a high-K+ diet. A possible role for K+ in chronic kidney disease is also emerging, as epidemiological data reveal associations between higher urinary K+ excretion and improved renal outcomes. This comprehensive review will embed these novel insights on NCC regulation into existing concepts of K+ homeostasis in health and disease.
Collapse
Affiliation(s)
- Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Martin Gritter
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Catherina A. Cuevas
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Robert A. Fenton
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|