1
|
Drzymała-Czyż S, Walkowiak J, Colombo C, Alicandro G, Storrösten OT, Kolsgaard M, Bakkeheim E, Strandvik B. Fatty acid abnormalities in cystic fibrosis-the missing link for a cure? iScience 2024; 27:111153. [PMID: 39620135 PMCID: PMC11607544 DOI: 10.1016/j.isci.2024.111153] [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: 01/05/2025] Open
Abstract
The care for cystic fibrosis (CF) has dramatically changed with the development of modulators, correctors, and potentiators of the CFTR molecule, which lead to improved clinical status of most people with CF (pwCF). The modulators influence phospholipids and ceramides, but not linoleic acid (LA) deficiency, associated with more severe phenotypes of CF. The LA deficiency is associated with upregulation of its transfer to arachidonic acid (AA). The AA release from membranes is increased and associated with increase of pro-inflammatory prostanoids and the characteristic inflammation is present before birth and bacterial infections. Docosahexaenoic acid is often decreased, especially in associated liver disease Some endogenously synthesized fatty acids are increased. Cholesterol and ceramide metabolisms are disturbed. The lipid abnormalities are present at birth, and before feeding in transgenic pigs and ferrets. This review focus on the lipid abnormalities and their associations to clinical symptoms in CF, based on clinical studies and experimental research.
Collapse
Affiliation(s)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Institute of Pediatrics, Poznan University of Medical Sciences, Poznan, Poland
| | - Carla Colombo
- Cystic Fibrosis Centre, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gianfranco Alicandro
- Cystic Fibrosis Centre, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Olav Trond Storrösten
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | - Magnhild Kolsgaard
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | - Egil Bakkeheim
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
2
|
Ma X, Xu R, Chen J, Wang S, Hu P, Wu Y, Que Y, Du W, Cai X, Chen H, Guo J, Li TC, Ruan YC. The epithelial Na + channel (ENaC) in ovarian granulosa cells modulates Ca 2+ mobilization and gonadotrophin signaling for estrogen homeostasis and female fertility. Cell Commun Signal 2024; 22:398. [PMID: 39143495 PMCID: PMC11323461 DOI: 10.1186/s12964-024-01778-5] [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: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
Ovarian granulosa cells are essential to gonadotrophin-regulated estrogen production, female cycle maintenance and fertility. The epithelial Na+ channel (ENaC) is associated with female fertility; however, whether and how it plays a role in ovarian cell function(s) remained unexplored. Here, we report patch-clamp and Na+ imaging detection of ENaC expression and channel activity in both human and mouse ovarian granulosa cells, which are promoted by pituitary gonadotrophins, follicle stimulating hormone (FSH) or luteinizing hormone (LH). Cre-recombinase- and CRISPR-Cas9-based granulosa-specific knockout of ENaC α subunit (Scnn1a) in mice resulted in failed estrogen elevation at early estrus, reduced number of corpus luteum, abnormally extended estrus phase, reduced litter size and subfertility in adult female mice. Further analysis using technologies including RNA sequencing and Ca2+ imaging revealed that pharmacological inhibition, shRNA-based knockdown or the knockout of ENaC diminished spontaneous or stimulated Ca2+ oscillations, lowered the capacity of intracellular Ca2+ stores and impaired FSH/LH-stimulated transcriptome changes for estrogen production in mouse and/or human granulosa cells. Together, these results have revealed a previously undefined role of ENaC in modulating gonadotrophin signaling in granulosa cells for estrogen homeostasis and thus female fertility.
Collapse
Affiliation(s)
- Xiyang Ma
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Ruiyao Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Junjiang Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Jinan University, Guangzhou, China
| | - Shan Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Peijie Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yong Wu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yanting Que
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wanting Du
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaojun Cai
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hui Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Tin Chiu Li
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China.
| |
Collapse
|
3
|
Moreira da Silva AE, Franco AM, Ferguson BS, Fonseca MA. Influence of previous plane of nutrition on molecular mechanisms regulating the expression of urea and water metabolism related genes in the rumen and kidney of finishing crossbred Angus steers. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:232-243. [PMID: 38800739 PMCID: PMC11126772 DOI: 10.1016/j.aninu.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 05/29/2024]
Abstract
This study aimed to understand how molecular mechanisms controlling water and urea metabolism at the finishing phase can be affected by previous plane of nutrition of crossbred Angus beef steers. Twenty-four (n = 24) animals were randomly distributed into either a moderate (MP) or high plane of nutrition during the background phase for 85 d. Animals were then blocked by their previous plane and were moved onto a 105-d finishing phase in a 2 × 2 factorial arrangement. The forage-finished group received only high-quality alfalfa hay, whereas the grain-fed group received a high grain diet (80% whole corn and 20% alfalfa hay). By the end of the finishing phase, animals were harvested and tissue samples from the rumen and kidney were collected. Changes in gene expression of aquaporins (AQP)-2, -3, -4, -7, ATP1A1, ATP1B1, SGK1, CLIC1 (kidney and rumen), UT-A1 (kidney only) and UT-B (rumen only), were assayed via real-time qPCR; 18S rRNA was used as an endogenous control. One-way ANOVA followed by Tukey's post hoc analysis was conducted. When animals were from MP, forage-finishing increased the relative abundance of AQP3 (P ≤ 0.05), AQP7 (P ≤ 0.05), ATP1B1 (P ≤ 0.05), and SGK1 (P ≤ 0.05) in the kidney when compared to grain-fed animals. In the rumen, for the MP group, AQP7 was differentially expressed in both treatments at the finishing phase (P ≤ 0.01), with forage-finished steers having the highest expression of AQP7. For the MP group, UT-B had a tendency of presenting a higher expression on grain-fed animals (P = 0.075). Overall, these results suggest that previous plane can impact expression of genes associated with water and urea metabolism during the finishing phase, namely AQP3, AQP7, ATP1B1, and SGK1 in the kidney, and AQP7 and UT-B in the rumen. The greatest impact observed on gene expression changes of investigated genes at the finishing phase was reflective of animals backgrounded on the moderate previous plane.
Collapse
Affiliation(s)
- Aghata E. Moreira da Silva
- College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, Reno, NV 89503, USA
| | - Arturo Macias Franco
- College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, Reno, NV 89503, USA
| | - Bradley S. Ferguson
- College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, Reno, NV 89503, USA
| | - Mozart A. Fonseca
- College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, Reno, NV 89503, USA
- Department of Animal and Range Sciences, Clayton Livestock Research Center, New Mexico State University, Clayton, NM 88415, USA
| |
Collapse
|
4
|
Sun K, Wang YL, Hou CC, Shang D, Du LJ, Bai L, Zhang XY, Hao CM, Duan SZ. Collecting duct NCOR1 controls blood pressure by regulating mineralocorticoid receptor. J Adv Res 2024:S2090-1232(24)00053-5. [PMID: 38341030 DOI: 10.1016/j.jare.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/29/2023] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Nuclear receptor corepressor 1(NCOR1) is reported to play crucial roles in cardiovascular diseases, but its function in the kidney has remained obscure. OBJECTIVE We aim to elucidate the role of collecting duct NCOR1 in blood pressure (BP) regulation. METHODS AND RESULTS Collecting duct NCOR1 knockout (KO) mice manifested increased BP and aggravated vascular and renal injury in an angiotensin II (Ang II)-induced hypertensive model. KO mice also showed significantly higher BP than littermate control (LC) mice in deoxycorticosterone acetate (DOCA)-salt model. Further study showed that collecting duct NCOR1 deficiency aggravated volume and sodium retention after saline challenge. Among the sodium transporter in the collecting duct, the expression of the three epithelial sodium channel (ENaC) subunits was markedly increased in the renal medulla of KO mice. Consistently, BP in Ang II-infused KO mice decreased significantly to the similar level as those in LC mice after amiloride treatment. ChIP analysis revealed that NCOR1 deficiency increased the enrichment of mineralocorticoid receptor (MR) on the promoters of the three ENaC genes in primary inner medulla collecting duct (IMCD) cells. Co-IP results showed interaction between NCOR1 and MR, and luciferase reporter results demonstrated that NCOR1 inhibited the transcriptional activity of MR. Knockdown of MR eliminated the increased ENaC expression in primary IMCD cells isolated from KO mice. Finally, BP was significantly decreased in Ang II-infused KO mice after treatment of MR antagonist spironolactone and the difference between LC and KO mice was abolished. CONCLUSIONS NCOR1 interacts with MR to control ENaC activity in the collecting duct and to regulate sodium reabsorption and ultimately BP. Targeting NCOR1 might be a promising tactic to interrupt the volume and sodium retention of the collecting duct in hypertension.
Collapse
Affiliation(s)
- Ke Sun
- Department of Nephrology, Zhejiang University Medical College Affiliated Sir Run Run Shaw Hospital, Hangzhou, Zhejiang Province 310016, China; Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yong-Li Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen-Chen Hou
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Da Shang
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lan Bai
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xing-Yu Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Sheng-Zhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine; State Key Laboratory of Transvascular Implantation Devices, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
5
|
Nethathe GD, Lipman J, Anderson R, Fuller PJ, Feldman C. Glucocorticoids with or without fludrocortisone in septic shock: a narrative review from a biochemical and molecular perspective. Br J Anaesth 2024; 132:53-65. [PMID: 38030548 PMCID: PMC10797514 DOI: 10.1016/j.bja.2023.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Two randomised controlled trials have reported a reduction in mortality when adjunctive hydrocortisone is administered in combination with fludrocortisone compared with placebo in septic shock. A third trial did not support this finding when hydrocortisone administered in combination with fludrocortisone was compared with hydrocortisone alone. The underlying mechanisms for this mortality benefit remain poorly understood. We review the clinical implications and potential mechanisms derived from laboratory and clinical data underlying the beneficial role of adjunctive fludrocortisone with hydrocortisone supplementation in septic shock. Factors including distinct biological effects of glucocorticoids and mineralocorticoids, tissue-specific and mineralocorticoid receptor-independent effects of mineralocorticoids, and differences in downstream signalling pathways between mineralocorticoid and glucocorticoid binding at the mineralocorticoid receptor could contribute to this interaction. Furthermore, pharmacokinetic and pharmacodynamic disparities exist between aldosterone and its synthetic counterpart fludrocortisone, potentially influencing their effects. Pending publication of well-designed, randomised controlled trials, a molecular perspective offers valuable insights and guidance to help inform clinical strategies.
Collapse
Affiliation(s)
- Gladness D Nethathe
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; Academy of Critical Care, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Jeffrey Lipman
- Academy of Critical Care, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia; Jamieson Trauma Institute and Intensive Care Services, Royal Brisbane and Women's Hospital, Butterfield Street, Herston, Brisbane, 4029, QLD, Australia; Nimes University Hospital, University of Montpellier, Nimes, France
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Peter J Fuller
- Endocrinology Unit, Monash Health, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Charles Feldman
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
6
|
Dasargyri A, González Rodríguez D, Rehrauer H, Reichmann E, Biedermann T, Moehrlen U. scRNA-Seq of Cultured Human Amniotic Fluid from Fetuses with Spina Bifida Reveals the Origin and Heterogeneity of the Cellular Content. Cells 2023; 12:1577. [PMID: 37371048 DOI: 10.3390/cells12121577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Amniotic fluid has been proposed as an easily available source of cells for numerous applications in regenerative medicine and tissue engineering. The use of amniotic fluid cells in biomedical applications necessitates their unequivocal characterization; however, the exact cellular composition of amniotic fluid and the precise tissue origins of these cells remain largely unclear. Using cells cultured from the human amniotic fluid of fetuses with spina bifida aperta and of a healthy fetus, we performed single-cell RNA sequencing to characterize the tissue origin and marker expression of cultured amniotic fluid cells at the single-cell level. Our analysis revealed nine different cell types of stromal, epithelial and immune cell phenotypes, and from various fetal tissue origins, demonstrating the heterogeneity of the cultured amniotic fluid cell population at a single-cell resolution. It also identified cell types of neural origin in amniotic fluid from fetuses with spina bifida aperta. Our data provide a comprehensive list of markers for the characterization of the various progenitor and terminally differentiated cell types in cultured amniotic fluid. This study highlights the relevance of single-cell analysis approaches for the characterization of amniotic fluid cells in order to harness their full potential in biomedical research and clinical applications.
Collapse
Affiliation(s)
- Athanasia Dasargyri
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, 8032 Zurich, Switzerland
| | - Daymé González Rodríguez
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057 Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057 Zurich, Switzerland
| | - Ernst Reichmann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, 8032 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Zurich Center for Fetal Diagnosis and Therapy, University of Zurich, 8006 Zurich, Switzerland
- Pediatric Surgery, University Children's Hospital Zurich, 8032 Zurich, Switzerland
| |
Collapse
|
7
|
Gonsalez SR, Gomes DS, de Souza AM, Ferrão FM, Vallotton Z, Gogulamudi VR, Lowe J, Casarini DE, Prieto MC, Lara LS. The Triad Na + Activated Na + Channel (Nax)-Salt Inducible KINASE (SIK) and (Na + + K +)-ATPase: Targeting the Villains to Treat Salt Resistant and Sensitive Hypertension. Int J Mol Sci 2023; 24:ijms24097887. [PMID: 37175599 PMCID: PMC10178781 DOI: 10.3390/ijms24097887] [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: 02/03/2023] [Revised: 04/04/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The Na+-activated Na+ channel (Nax) and salt-inducible kinase (SIK) are stimulated by increases in local Na+ concentration, affecting (Na+ + K+)-ATPase activity. To test the hypothesis that the triad Nax/SIK/(Na+ + K+)-ATPase contributes to kidney injury and salt-sensitive hypertension (HTN), uninephrectomized male Wistar rats (200 g; n = 20) were randomly divided into 4 groups based on a salt diet (normal salt diet; NSD-0.5% NaCl-or high-salt diet; HSD-4% NaCl) and subcutaneous administration of saline (0.9% NaCl) or deoxycorticosterone acetate (DOCA, 8 mg/kg), as follows: Control (CTRL), CTRL-Salt, DOCA, and DOCA-Salt, respectively. After 28 days, the following were measured: kidney function, blood pressure, (Na+ + K+)-ATPase and SIK1 kidney activities, and Nax and SIK1 renal expression levels. SIK isoforms in kidneys of CTRL rats were present in the glomerulus and tubular epithelia; they were not altered by HSD and/or HTN. CTRL-Salt rats remained normotensive but presented slight kidney function decay. HSD rats displayed augmentation of the Nax/SIK/(Na+ + K+)-ATPase pathway. HTN, kidney injury, and kidney function decay were present in all DOCA rats; these were aggravated by HSD. DOCA rats presented unaltered (Na+ + K+)-ATPase activity, diminished total SIK activity, and augmented SIK1 and Nax content in the kidney cortex. DOCA-Salt rats expressed SIK1 activity and downregulation in (Na+ + K+)-ATPase activity in the kidney cortex despite augmented Nax content. The data of this study indicate that the (Na+ + K+)-ATPase activity response to SIK is attenuated in rats under HSD, independent of HTN, as a mechanism contributing to kidney injury and salt-sensitive HTN.
Collapse
Affiliation(s)
- Sabrina R Gonsalez
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Campus Macaé, Rio de Janeiro 21941-901, Brazil
| | - Dayene S Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-971, Brazil
| | - Alessandro M de Souza
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-971, Brazil
| | - Fernanda M Ferrão
- Núcleo Multidisciplinar de Pesquisa em Biologia (NUMPEX-BIO), Universidade Federal do Rio de Janeiro, Campus Caxias, Rio de Janeiro 21941-901, Brazil
| | - Zoe Vallotton
- Department of Physiology, School of Medicine and Tulane Renal and Hypertension Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Venkateswara R Gogulamudi
- Department of Physiology, School of Medicine and Tulane Renal and Hypertension Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jennifer Lowe
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Dulce E Casarini
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Minolfa C Prieto
- Department of Physiology, School of Medicine and Tulane Renal and Hypertension Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lucienne S Lara
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-971, Brazil
| |
Collapse
|
8
|
Noroozbabaee L, Blanco PJ, Safaei S, Nickerson DP. A modular and reusable model of epithelial transport in the proximal convoluted tubule. PLoS One 2022; 17:e0275837. [PMID: 36355848 PMCID: PMC9648790 DOI: 10.1371/journal.pone.0275837] [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: 01/16/2022] [Accepted: 09/24/2022] [Indexed: 11/12/2022] Open
Abstract
We review a collection of published renal epithelial transport models, from which we build a consistent and reusable mathematical model able to reproduce many observations and predictions from the literature. The flexible modular model we present here can be adapted to specific configurations of epithelial transport, and in this work we focus on transport in the proximal convoluted tubule of the renal nephron. Our mathematical model of the epithelial proximal convoluted tubule describes the cellular and subcellular mechanisms of the transporters, intracellular buffering, solute fluxes, and other processes. We provide free and open access to the Python implementation to ensure our multiscale proximal tubule model is accessible; enabling the reader to explore the model through setting their own simulations, reproducibility tests, and sensitivity analyses.
Collapse
Affiliation(s)
- Leyla Noroozbabaee
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Pablo J. Blanco
- National Laboratory for Scientific Computing, Petrópolis, Brazil
| | - Soroush Safaei
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - David P. Nickerson
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 0] [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
|
10
|
Mohammadi S, Yang L, Bulbert M, Rowland HM. Defence mitigation by predators of chemically defended prey integrated over the predation sequence and across biological levels with a focus on cardiotonic steroids. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220363. [PMID: 36133149 PMCID: PMC9449480 DOI: 10.1098/rsos.220363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/17/2022] [Indexed: 05/10/2023]
Abstract
Predator-prey interactions have long served as models for the investigation of adaptation and fitness in natural environments. Anti-predator defences such as mimicry and camouflage provide some of the best examples of evolution. Predators, in turn, have evolved sensory systems, cognitive abilities and physiological resistance to prey defences. In contrast to prey defences which have been reviewed extensively, the evolution of predator counter-strategies has received less attention. To gain a comprehensive view of how prey defences can influence the evolution of predator counter-strategies, it is essential to investigate how and when selection can operate. In this review we evaluate how predators overcome prey defences during (i) encounter, (ii) detection, (iii) identification, (iv) approach, (v) subjugation, and (vi) consumption. We focus on prey that are protected by cardiotonic steroids (CTS)-defensive compounds that are found in a wide range of taxa, and that have a specific physiological target. In this system, coevolution is well characterized between specialist insect herbivores and their host plants but evidence for coevolution between CTS-defended prey and their predators has received less attention. Using the predation sequence framework, we organize 574 studies reporting predators overcoming CTS defences, integrate these counter-strategies across biological levels of organization, and discuss the costs and benefits of attacking CTS-defended prey. We show that distinct lineages of predators have evolved dissecting behaviour, changes in perception of risk and of taste perception, and target-site insensitivity. We draw attention to biochemical, hormonal and microbiological strategies that have yet to be investigated as predator counter-adaptations to CTS defences. We show that the predation sequence framework will be useful for organizing future studies of chemically mediated systems and coevolution.
Collapse
Affiliation(s)
- Shabnam Mohammadi
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA
- Institut für Zell- und Systembiologie der Tiere, Universität Hamburg, Hamburg, Germany
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Lu Yang
- Wellcome Sanger Institute, Cambridge, UK
| | - Matthew Bulbert
- Department of Biological Sciences, Macquarie University North Ryde, New South Wales, Australia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, University of Oxford Brookes, Oxford, UK
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | | |
Collapse
|
11
|
Mohammadi S, Yang L, Bulbert M, Rowland HM. Defence mitigation by predators of chemically defended prey integrated over the predation sequence and across biological levels with a focus on cardiotonic steroids. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220363. [PMID: 36133149 DOI: 10.6084/m9.figshare.c.6168216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/17/2022] [Indexed: 05/25/2023]
Abstract
Predator-prey interactions have long served as models for the investigation of adaptation and fitness in natural environments. Anti-predator defences such as mimicry and camouflage provide some of the best examples of evolution. Predators, in turn, have evolved sensory systems, cognitive abilities and physiological resistance to prey defences. In contrast to prey defences which have been reviewed extensively, the evolution of predator counter-strategies has received less attention. To gain a comprehensive view of how prey defences can influence the evolution of predator counter-strategies, it is essential to investigate how and when selection can operate. In this review we evaluate how predators overcome prey defences during (i) encounter, (ii) detection, (iii) identification, (iv) approach, (v) subjugation, and (vi) consumption. We focus on prey that are protected by cardiotonic steroids (CTS)-defensive compounds that are found in a wide range of taxa, and that have a specific physiological target. In this system, coevolution is well characterized between specialist insect herbivores and their host plants but evidence for coevolution between CTS-defended prey and their predators has received less attention. Using the predation sequence framework, we organize 574 studies reporting predators overcoming CTS defences, integrate these counter-strategies across biological levels of organization, and discuss the costs and benefits of attacking CTS-defended prey. We show that distinct lineages of predators have evolved dissecting behaviour, changes in perception of risk and of taste perception, and target-site insensitivity. We draw attention to biochemical, hormonal and microbiological strategies that have yet to be investigated as predator counter-adaptations to CTS defences. We show that the predation sequence framework will be useful for organizing future studies of chemically mediated systems and coevolution.
Collapse
Affiliation(s)
- Shabnam Mohammadi
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA
- Institut für Zell- und Systembiologie der Tiere, Universität Hamburg, Hamburg, Germany
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Lu Yang
- Wellcome Sanger Institute, Cambridge, UK
| | - Matthew Bulbert
- Department of Biological Sciences, Macquarie University North Ryde, New South Wales, Australia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, University of Oxford Brookes, Oxford, UK
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | | |
Collapse
|
12
|
Glucocorticoids Promote Na+ Excretion in the Renal Epithelia of Heart Failure Rats by Suppressing Transporter Proteins Involved in Acute Sodium Loading. J Cardiovasc Pharmacol 2022; 80:453-463. [PMID: 35853190 DOI: 10.1097/fjc.0000000000001310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/16/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Glucocorticoid receptors are essential for normal development and stress responses. Their role in H 2 O and Na + metabolism, especially in chronic heart failure (CHF), is not well defined. In a previous study, we found that glucocorticoids potentiate urination in CHF and promote H 2 O excretion by inhibiting the vasopressin receptor 2 pathway. The present study examines the effect of glucocorticoids on renal Na + excretion and the underlying mechanisms in CHF rats with acute sodium loading. CHF was induced by left coronary artery ligation for 8 weeks. Rats were randomly assigned to 5 groups: control, CHF, dexamethasone (DEX)-administered CHF, DEX-administered CHF treated with RU486 (mifepristone, a glucocorticoid receptor antagonist), and RU486-treated CHF. An acute sodium loading test was performed 6 hours after DEX administration. Blood and urine samples were collected, and hemodynamics were measured. The expression and localization of Na + transporter proteins were determined by immunoblotting and immunohistochemistry. DEX increased the urine volume and urinary sodium and improved cardiac function and the estimated glomerular filtration rate in CHF rats. The upregulation of the epithelial sodium channel β and γ subunits, Na-K-2Cl cotransporter, serum glucocorticoid-regulated kinase 1 (SGK1), and Na + /K + -ATPase in the renal epithelium of CHF rats was downregulated by DEX. These beneficial effects were abolished by RU486. The expression of natriuretic peptide receptor A was opposite that of the above proteins. Glucocorticoids might induce profound natriuresis in CHF rats during acute sodium loading, which is associated with downregulating some Na + transporter proteins in the renal epithelium and improving intrarenal hemodynamics.
Collapse
|
13
|
Beverley KM, Pattnaik BR. Inward rectifier potassium (Kir) channels in the retina: living our vision. Am J Physiol Cell Physiol 2022; 323:C772-C782. [PMID: 35912989 PMCID: PMC9448332 DOI: 10.1152/ajpcell.00112.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022]
Abstract
Channel proteins are vital for conducting ions throughout the body and are especially relevant to retina physiology. Inward rectifier potassium (Kir) channels are a class of K+ channels responsible for maintaining membrane potential and extracellular K+ concentrations. Studies of the KCNJ gene (that encodes Kir protein) expression identified the presence of all of the subclasses (Kir 1-7) of Kir channels in the retina or retinal-pigmented epithelium (RPE). However, functional studies have established the involvement of the Kir4.1 homotetramer and Kir4.1/5.1 heterotetramer in Müller glial cells, Kir2.1 in bipolar cells, and Kir7.1 in the RPE cell physiology. Here, we propose the potential roles of Kir channels in the retina based on the physiological contributions to the brain, pancreatic, and cardiac tissue functions. There are several open questions regarding the expressed KCNJ genes in the retina and RPE. For example, why does not the Kir channel subtype gene expression correspond with protein expression? Catching up with multiomics or functional "omics" approaches might shed light on posttranscriptional changes that might influence Kir subunit mRNA translation within the retina that guides our vision.
Collapse
Affiliation(s)
- Katie M Beverley
- Endocrinology and Reproductive Physiology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Bikash R Pattnaik
- Endocrinology and Reproductive Physiology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
14
|
Rodan AR. Regulation of Distal Nephron Transport by Intracellular Chloride and Potassium. Nephron Clin Pract 2022; 147:203-211. [PMID: 35977527 PMCID: PMC9935751 DOI: 10.1159/000526051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Low potassium increases the phosphorylation and activity of the sodium chloride cotransporter (NCC) in the distal convoluted tubule of the nephron, which contributes to the hypertensive effect of the modern low potassium/high sodium diet. A central mediator of potassium regulation of NCC is the chloride-sensitive With No Lysine [K] (WNK) kinase. SUMMARY Chloride directly inhibits WNKs by binding to the active site. The mechanisms underlying WNK regulation by extracellular potassium are reviewed, as well as the modulatory effect of kidney-specific-WNK1. WNK1, but not WNK1 kinase activity, is also required for the aldosterone-independent regulation of the epithelial sodium channel by potassium. Whether intracellular chloride could be involved in this process is discussed. Recent studies demonstrating direct regulation of WNKs by intracellular potassium are also reviewed, and the potential physiological relevance to renal epithelial ion transport is discussed. KEY MESSAGES WNKs are sensors of the intracellular ionic milieu. In the nephron, changes in extracellular ion concentrations, resulting in changes in intracellular ion concentration, regulate WNK activity and downstream transporters and channels to maintain total body ion homeostasis.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| |
Collapse
|
15
|
Renal water transport in health and disease. Pflugers Arch 2022; 474:841-852. [PMID: 35678906 PMCID: PMC9338902 DOI: 10.1007/s00424-022-02712-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
Abstract
Saving body water by optimal reabsorption of water filtered by the kidney leading to excretion of urine with concentrations of solutes largely above that of plasma allowed vertebrate species to leave the aquatic environment to live on solid ground. Filtered water is reabsorbed for 70% and 20% by proximal tubules and thin descending limbs of Henle, respectively. These two nephron segments express the water channel aquaporin-1 located along both apical and basolateral membranes. In the proximal tubule, the paracellular pathway accounts for at least 30% of water reabsorption, and the tight-junction core protein claudin-2 plays a key role in this permeability. The ascending limb of Henle and the distal convoluted tubule are impermeant to water and are responsible for urine dilution. The water balance is adjusted along the collecting system, i.e. connecting tubule and the collecting duct, under the control of arginine-vasopressin (AVP). AVP is synthesized by the hypothalamus and released in response to an increase in extracellular osmolality or stimulation of baroreceptors by decreased blood pressure. In response to AVP, aquaporin-2 water channels stored in subapical intracellular vesicles are translocated to the apical plasma membrane and raise the water permeability of the collecting system. The basolateral step of water reabsorption is mediated by aquaporin-3 and -4, which are constitutively expressed. Drugs targeting water transport include classical diuretics, which primarily inhibit sodium transport; the new class of SGLT2 inhibitors, which promotes osmotic diuresis and the non-peptidic antagonists of the V2 receptor, which are pure aquaretic drugs. Disturbed water balance includes diabetes insipidus and hyponatremias. Diabetes insipidus is characterized by polyuria and polydipsia. It is either related to a deficit in AVP secretion called central diabetes insipidus that can be treated by AVP analogs or to a peripheral defect in AVP response called nephrogenic diabetes insipidus. Diabetes insipidus can be either of genetic origin or acquired. Hyponatremia is a common disorder most often related to free water excess relying on overstimulated or inappropriate AVP secretion. The assessment of blood volume is key for the diagnosis and treatment of hyponatremia, which can be classified as hypo-, eu-, or hypervolemic.
Collapse
|
16
|
Hua Y, Han A, Yu T, Hou Y, Ding Y, Nie H. Small Extracellular Vesicles Containing miR-34c Derived from Bone Marrow Mesenchymal Stem Cells Regulates Epithelial Sodium Channel via Targeting MARCKS. Int J Mol Sci 2022; 23:ijms23095196. [PMID: 35563590 PMCID: PMC9101277 DOI: 10.3390/ijms23095196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/18/2022] Open
Abstract
Epithelial sodium channel (ENaC) is a pivotal regulator of alveolar fluid clearance in the airway epithelium and plays a key role in the treatment of acute lung injury (ALI), which is mainly composed of the three homologous subunits (α, β and γ). The mechanisms of microRNAs in small extracellular vesicles (sEVs) derived from mesenchymal stem cell (MSC-sEVs) on the regulation of lung ion transport are seldom reported. In this study, we aimed at investigating whether miR-34c had an effect on ENaC dysfunction induced by lipopolysaccharide and explored the underlying mechanism in this process. Primarily, the effect of miR-34c on lung edema and histopathology changes in an ALI mouse model was investigated. Then the uptake of PKH26-labeled sEVs was observed in recipient cells, and we observed that the overexpression of miR-34c in MSC-sEVs could upregulate the LPS-inhibited γ-ENaC expression. The dual luciferase reporter gene assay demonstrated that myristoylated alanine-rich C kinase substrate (MARCKS) was one of target genes of miR-34c, the protein expression of which was negatively correlated with miR-34c. Subsequently, either upregulating miR-34c or knocking down MARCKS could increase the protein expression of phospho-phosphatidylinositol 3-kinase (p-PI3K) and phospho-protein kinase B (p-AKT), implying a downstream regulation pathway was involved. All of the above suggest that miR-34c in MSC-sEVs can attenuate edematous lung injury via enhancing γ-ENaC expression, at least partially, through targeting MARCKS and activating the PI3K/AKT signaling pathway subsequently.
Collapse
|
17
|
Khundmiri SJ, Ecelbarger CM, Amponsem J, Ji H, Sandberg K, Lee DL. PPAR-α knockout leads to elevated blood pressure response to angiotensin II infusion associated with an increase in renal α-1 Na +/K + ATPase protein expression and activity. Life Sci 2022; 296:120444. [PMID: 35245523 PMCID: PMC8969884 DOI: 10.1016/j.lfs.2022.120444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 02/15/2022] [Accepted: 02/26/2022] [Indexed: 01/13/2023]
Abstract
Peroxisome proliferator activated receptor alpha (PPAR-α) deletion has been shown to increase blood pressure (BP). We hypothesized that the BP increase in PPAR-α KO mice was mediated by increased expression and activity of basolateral Na+/K+ ATPase (NKA) pump. To address this hypothesis, we treated wild-type (WT) and PPAR-α knockout (KO) mice with a slow-pressor dose of angiotensin II (400 ng/kg·min) for 12 days by osmotic minipump. Radiotelemetry showed no significant differences in baseline mean arterial pressure (MAP) between WT and PPAR-α KO mice; however, by day 12 of infusion, MAP was significantly higher in PPAR-α KO mice (156 ± 16) compared to WT mice (138 ± 11 mmHg). NKA activity and protein expression (α1 subunit) were significantly higher in PPAR-α KO mice compared to WT mice. There was no significant difference in NKA mRNA levels. Angiotensin II further increased the expression and activity of the NKA in both genotypes along with the water channel, aquaporin 1 (Aqp1). In contrast, angiotensin II decreased the expression (64-97% reduction in band density) of sodium‑hydrogen exchanger-3 (NHE3), NHE regulatory factor-1 (NHERF1, Slc9a3r1), sodium‑potassium-2-chloride cotransporter (NKCC2), and epithelial sodium channel (ENaC) β- and γ- subunits in the renal cortex of both WT and PPAR-α KO mice, with no difference between genotypes. The sodium-chloride cotransporter (NCC) was also decreased by angiotensin II, but significantly more in PPAR-α KO (59% WT versus 77% KO reduction from their respective vehicle-treated mice). Our results suggest that PPAR-α attenuates angiotensin II-mediated increased blood pressure potentially via reducing expression and activity of the NKA.
Collapse
Affiliation(s)
- Syed J Khundmiri
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA.
| | - Carolyn M Ecelbarger
- Division of Endocrinology and Metabolism, Department of Medicine, Georgetown University Washington, DC, USA
| | - Joycemary Amponsem
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Hong Ji
- Department of Medicine, Georgetown University Washington, DC, USA
| | - Kathryn Sandberg
- Department of Medicine, Georgetown University Washington, DC, USA
| | - Dexter L Lee
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA.
| |
Collapse
|
18
|
Sudarikova AV, Fomin MV, Sultanova RF, Zhao Y, Perez S, Domondon M, Shamatova M, Lysikova DV, Spires DR, Ilatovskaya DV. Functional role of histamine receptors in the renal cortical collecting duct cells. Am J Physiol Cell Physiol 2022; 322:C775-C786. [PMID: 35081320 PMCID: PMC8993525 DOI: 10.1152/ajpcell.00420.2021] [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: 11/22/2022]
Abstract
Histamine is an important immunomodulator, as well as a regulator of allergic inflammation, gastric acid secretion, and neurotransmission. Although substantial histamine level has been reported in the kidney, renal pathological and physiological effects of this compound have not been clearly defined. The goal of this study was to provide insight into the role of histamine-related pathways in the kidney, with emphasis on the collecting duct (CD), a distal part of the nephron important for the regulation of blood pressure. We report that all four histamine receptors (HRs) as well as enzymes responsible for histamine metabolism and synthesis are expressed in cultured mouse mpkCCDcl4 cells, and histamine evokes a dose-dependent transient increase in intracellular Ca2+ in these cells. Furthermore, we observed a dose-dependent increase in cAMP in the CD cells in response to histamine. Short-circuit current studies aimed at measuring Na+ reabsorption via ENaC (epithelial Na+ channel) demonstrated inhibition of ENaC-mediated currents by histamine after a 4-hr incubation, and single-channel patch-clamp analysis revealed similar ENaC open probability before and after acute histamine application. The long-term (4 hr) effect on ENaC was corroborated in immunocytochemistry and qPCR, which showed a decrease in protein and gene expression for αENaC upon histamine treatment. In summary, our data highlight the functional importance of HRs in the CD cells and suggest potential implications of histamine in inflammation-related renal conditions. Further research is required to discern the molecular pathways downstream of HRs and assess the role of specific receptors in renal pathophysiology.
Collapse
Affiliation(s)
- Anastasia V Sudarikova
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia, St. Petersburg
| | - Mikhail V Fomin
- Department of Physiology, Augusta University, Augusta, United States
| | - Regina F Sultanova
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Charleston, United States
| | - Ying Zhao
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Charleston, United States
| | - Samantha Perez
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Charleston, United States
| | - Mark Domondon
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Charleston, United States
| | - Margarita Shamatova
- grid.410427.4Augusta University (Augusta, Georgia, United States), Augusta, United States
| | - Daria V Lysikova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia; Department of Physiology, Augusta University, United States, Augusta, United States
| | - Denisha R Spires
- Department of Physiology, Augusta University, Augusta, Georgia, United States
| | | |
Collapse
|
19
|
Jiang C, Xie S, Yang G, Wang N. Spotlight on NLRP3 Inflammasome: Role in Pathogenesis and Therapies of Atherosclerosis. J Inflamm Res 2022; 14:7143-7172. [PMID: 34992411 PMCID: PMC8711145 DOI: 10.2147/jir.s344730] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an intricate biological response of body tissues to detrimental stimuli. Cardiovascular disease (CVD) is the leading cause of death worldwide, and inflammation is well documented to play a role in the development of CVD, especially atherosclerosis (AS). Emerging evidence suggests that activation of the NOD-like receptor (NLR) family and the pyridine-containing domain 3 (NLRP3) inflammasome is instrumental in inflammation and may result in AS. The NLRP3 inflammasome acts as a molecular platform that triggers the activation of caspase-1 and the cleavage of pro-interleukin (IL)-1β, pro-IL-18, and gasdermin D (GSDMD). The cleaved GSDMD forms pores in the cell membrane and initiates pyroptosis, inducing cell death and the discharge of intracellular pro-inflammatory factors. Hence, the NLRP3 inflammasome is a promising target for anti-inflammatory therapy against AS. In this review, we systematically summarized the current understanding of the activation mechanism of NLRP3 inflammasome, and the pathological changes in AS involving NLRP3. We also discussed potential therapeutic strategies targeting NLRP3 inflammasome to combat AS.
Collapse
Affiliation(s)
- Chunteng Jiang
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany
| | - Santuan Xie
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Guang Yang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Ningning Wang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
20
|
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
|
21
|
Marchini M, Ashkin MR, Bellini M, Sun MMG, Workentine ML, Okuyan HM, Krawetz R, Beier F, Rolian C. A Na +/K + ATPase Pump Regulates Chondrocyte Differentiation and Bone Length Variation in Mice. Front Cell Dev Biol 2022; 9:708384. [PMID: 34970538 PMCID: PMC8712571 DOI: 10.3389/fcell.2021.708384] [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: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022] Open
Abstract
The genetic and developmental mechanisms involved in limb formation are relatively well documented, but how these mechanisms are modulated by changes in chondrocyte physiology to produce differences in limb bone length remains unclear. Here, we used high throughput RNA sequencing (RNAseq) to probe the developmental genetic basis of variation in limb bone length in Longshanks, a mouse model of experimental evolution. We find that increased tibia length in Longshanks is associated with altered expression of a few key endochondral ossification genes such as Npr3, Dlk1, Sox9, and Sfrp1, as well reduced expression of Fxyd2, a facultative subunit of the cell membrane-bound Na+/K+ ATPase pump (NKA). Next, using murine tibia and cell cultures, we show a dynamic role for NKA in chondrocyte differentiation and in bone length regulation. Specifically, we show that pharmacological inhibition of NKA disrupts chondrocyte differentiation, by upregulating expression of mesenchymal stem cell markers (Prrx1, Serpina3n), downregulation of chondrogenesis marker Sox9, and altered expression of extracellular matrix genes (e.g., collagens) associated with proliferative and hypertrophic chondrocytes. Together, Longshanks and in vitro data suggest a broader developmental and evolutionary role of NKA in regulating limb length diversity.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Mitchell R Ashkin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Melina Bellini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew Lloyd Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hamza Malik Okuyan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Roman Krawetz
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Petrič M, Vidović A, Dolinar K, Miš K, Chibalin AV, Pirkmajer S. Phosphorylation of Na +,K +-ATPase at Tyr10 of the α1-Subunit is Suppressed by AMPK and Enhanced by Ouabain in Cultured Kidney Cells. J Membr Biol 2021; 254:531-548. [PMID: 34748042 PMCID: PMC8595181 DOI: 10.1007/s00232-021-00209-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023]
Abstract
Na+,K+-ATPase (NKA) is essential for maintenance of cellular and whole-body water and ion homeostasis. In the kidney, a major site of ion transport, NKA consumes ~ 50% of ATP, indicating a tight coordination of NKA and energy metabolism. AMP-activated protein kinase (AMPK), a cellular energy sensor, regulates NKA by modulating serine phosphorylation of the α1-subunit, but whether it modulates other important regulatory phosphosites, such as Tyr10, is unknown. Using human kidney (HK-2) cells, we determined that the phosphorylation of Tyr10 was stimulated by the epidermal growth factor (EGF), which was opposed by inhibitors of Src kinases (PP2), tyrosine kinases (genistein), and EGF receptor (EGFR, gefitinib). AMPK activators AICAR and A-769662 suppressed the EGF-stimulated phosphorylation of EGFR (Tyr1173) and NKAα1 at Tyr10. The phosphorylation of Src (Tyr416) was unaltered by AICAR and increased by A-769662. Conversely, ouabain (100 nM), a pharmacological NKA inhibitor and a putative adrenocortical hormone, enhanced the EGF-stimulated Tyr10 phosphorylation without altering the phosphorylation of EGFR (Tyr1173) or Src (Tyr416). Ouabain (100–1000 nM) increased the ADP:ATP ratio, while it suppressed the lactate production and the oxygen consumption rate in a dose-dependent manner. Treatment with ouabain or gene silencing of NKAα1 or NKAα3 subunit did not activate AMPK. In summary, AMPK activators and ouabain had antagonistic effects on the phosphorylation of NKAα1 at Tyr10 in cultured HK-2 cells, which implicates a role for Tyr10 in coordinated regulation of NKA-mediated ion transport and energy metabolism.
Collapse
Affiliation(s)
- Metka Petrič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Anja Vidović
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Dolinar
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Miš
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Alexander V Chibalin
- National Research Tomsk State University, Tomsk, Russia. .,Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
| | - Sergej Pirkmajer
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
23
|
Laulhé M, Dumeige L, Vu TA, Hani I, Pussard E, Lombès M, Viengchareun S, Martinerie L. Sexual Dimorphism of Corticosteroid Signaling during Kidney Development. Int J Mol Sci 2021; 22:ijms22105275. [PMID: 34069759 PMCID: PMC8155845 DOI: 10.3390/ijms22105275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
Sexual dimorphism involves differences between biological sexes that go beyond sexual characteristics. In mammals, differences between sexes have been demonstrated regarding various biological processes, including blood pressure and predisposition to develop hypertension early in adulthood, which may rely on early events during development and in the neonatal period. Recent studies suggest that corticosteroid signaling pathways (comprising glucocorticoid and mineralocorticoid signaling pathways) have distinct tissue-specific expression and regulation during this specific temporal window in a sex-dependent manner, most notably in the kidney. This review outlines the evidence for a gender differential expression and activation of renal corticosteroid signaling pathways in the mammalian fetus and neonate, from mouse to human, that may favor mineralocorticoid signaling in females and glucocorticoid signaling in males. Determining the effects of such differences may shed light on short term and long term pathophysiological consequences, markedly for males.
Collapse
Affiliation(s)
- Margaux Laulhé
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
| | - Laurence Dumeige
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
- Pediatric Endocrinology Department, Hôpital Universitaire Robert Debre, France & Université de Paris, 75019 Paris, France
| | - Thi An Vu
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
| | - Imene Hani
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
| | - Eric Pussard
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Assistance Publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Marc Lombès
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
| | - Say Viengchareun
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
| | - Laetitia Martinerie
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, CEDEX, 94276 Le Kremlin-Bicêtre, France; (M.L.); (L.D.); (T.A.V.); (I.H.); (E.P.); (M.L.); (S.V.)
- Pediatric Endocrinology Department, Hôpital Universitaire Robert Debre, France & Université de Paris, 75019 Paris, France
- Correspondence:
| |
Collapse
|
24
|
Sholokh A, Klussmann E. Local cyclic adenosine monophosphate signalling cascades-Roles and targets in chronic kidney disease. Acta Physiol (Oxf) 2021; 232:e13641. [PMID: 33660401 DOI: 10.1111/apha.13641] [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] [Received: 11/02/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022]
Abstract
The molecular mechanisms underlying chronic kidney disease (CKD) are poorly understood and treatment options are limited, a situation underpinning the need for elucidating the causative molecular mechanisms and for identifying innovative treatment options. It is emerging that cyclic 3',5'-adenosine monophosphate (cAMP) signalling occurs in defined cellular compartments within nanometre dimensions in processes whose dysregulation is associated with CKD. cAMP compartmentalization is tightly controlled by a specific set of proteins, including A-kinase anchoring proteins (AKAPs) and phosphodiesterases (PDEs). AKAPs such as AKAP18, AKAP220, AKAP-Lbc and STUB1, and PDE4 coordinate arginine-vasopressin (AVP)-induced water reabsorption by collecting duct principal cells. However, hyperactivation of the AVP system is associated with kidney damage and CKD. Podocyte injury involves aberrant AKAP signalling. cAMP signalling in immune cells can be local and slow the progression of inflammatory processes typical for CKD. A major risk factor of CKD is hypertension. cAMP directs the release of the blood pressure regulator, renin, from juxtaglomerular cells, and plays a role in Na+ reabsorption through ENaC, NKCC2 and NCC in the kidney. Mutations in the cAMP hydrolysing PDE3A that cause lowering of cAMP lead to hypertension. Another major risk factor of CKD is diabetes mellitus. AKAP18 and AKAP150 and several PDEs are involved in insulin release. Despite the increasing amount of data, an understanding of functions of compartmentalized cAMP signalling with relevance for CKD is fragmentary. Uncovering functions will improve the understanding of physiological processes and identification of disease-relevant aberrations may guide towards new therapeutic concepts for the treatment of CKD.
Collapse
Affiliation(s)
- Anastasiia Sholokh
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
| | - Enno Klussmann
- Max‐Delbrück‐Center for Molecular Medicine (MDC) Helmholtz Association Berlin Germany
- DZHK (German Centre for Cardiovascular Research) Berlin Germany
| |
Collapse
|
25
|
Li C, Chen M, He X, Ouyang D. A mini-review on ion fluxes that regulate NLRP3 inflammasome activation. Acta Biochim Biophys Sin (Shanghai) 2021; 53:131-139. [PMID: 33355638 DOI: 10.1093/abbs/gmaa155] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Indexed: 12/15/2022] Open
Abstract
The activation of NLR family pyrin domain containing 3 (NLRP3) inflammasome can be induced by a wide spectrum of activators. This is unlikely achieved by the binding of different activators directly to the NLRP3 protein itself, as the activators found so far show different forms of chemical structures. Previous studies have shown that these activators can induce potassium ion (K+) and chloride ion (Cl-) efflux, calcium (Ca2+) and other ion mobilization, mitochondrial dysfunction, and lysosomal disruption, all of which are believed to cause NLRP3 inflammasome activation; how these events are induced by the activators and how they coordinate with each other in inducing the NLRP3 inflammasome activation are not fully understood. Increasing evidence suggests that the coordinated change of intracellular ion concentrations may be a common mechanism for the NLRP3 activation by different activators. In this mini-review, we present a brief summary of the current knowledge about how different ionic flows (including K+, sodium ion, Ca2+, magnesium ion, manganese ion, zinc ion, iron ion, and Cl-) are involved in regulating the NLRP3 inflammasome activation in macrophages.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Mingye Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xianhui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
26
|
Abstract
The current paradigm of type 2 diabetes (T2D) is gluco-centric, being exclusively categorized by glycemic characteristics. The gluco-centric paradigm views hyperglycemia as the primary target, being driven by resistance to insulin combined with progressive beta cells failure, and considers glycemic control its ultimate treatment goal. Most importantly, the gluco-centric paradigm considers the non-glycemic diseases associated with T2D, e.g., obesity, dyslipidemia, hypertension, macrovascular disease, microvascular disease and fatty liver as 'risk factors' and/or 'outcomes' and/or 'comorbidities', rather than primary inherent disease aspects of T2D. That is in spite of their high prevalence (60-90%) and major role in profiling T2D morbidity and mortality. Moreover, the gluco-centric paradigm fails to realize that the non-glycemic diseases of T2D are driven by insulin and, except for glycemic control, response to insulin in T2D is essentially the rule rather than the exception. Failure of the gluco-centric paradigm to offer an exhaustive unifying view of the glycemic and non-glycemic diseases of T2D may have contributed to T2D being still an unmet need. An mTORC1-centric paradigm maintains that hyperactive mTORC1 drives the glycemic and non-glycemic disease aspects of T2D. Hyperactive mTORC1 is proposed to act as double-edged agent, namely, to interfere with glycemic control by disrupting the insulin receptor-Akt transduction pathway, while concomitantly driving the non-glycemic diseases of T2D. The mTORC1-centric paradigm may offer a novel perspective for T2D in terms of pathogenesis, clinical focus and treatment strategy.
Collapse
Affiliation(s)
- Jacob Bar-Tana
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
27
|
Abstract
Primary aldosteronism remains a leading cause of secondary hypertension, and its diagnosis and management continue to pose a challenge for clinicians. In this article, we review the diagnosis of primary aldosteronism along with its cardiovascular manifestations. Treatment is described depending on the diagnostic outcome, focusing on medical management with mineralocorticoid receptor antagonists and unilateral adrenalectomy. Although screening and diagnosing hyperaldosteronism follows well-known algorithms, in practice, physicians may find difficulty establishing the best course of action due to complexity in testing and confirming laterality of aldosterone production by the adrenals. Recognizing and treating primary aldosteronism requires a multidisciplinary approach with primary care physicians, cardiologists, endocrinologists, and radiologists working collaboratively.
Collapse
|
28
|
Zhu X, Huang Y, Li S, Ge N, Li T, Wang Y, Liu K, Liu C. Glucocorticoids Reverse Diluted Hyponatremia Through Inhibiting Arginine Vasopressin Pathway in Heart Failure Rats. J Am Heart Assoc 2020; 9:e014950. [PMID: 32390535 PMCID: PMC7660850 DOI: 10.1161/jaha.119.014950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Arginine vasopressin dependent antidiuresis plays a key role in water‐sodium retention in heart failure. In recent years, the role of glucocorticoids in the control of body fluid homeostasis has been extensively investigated. Glucocorticoid deficiency can activate V2R (vasopressin receptor 2), increase aquaporins expression, and result in hyponatremia, all of which can be reversed by glucocorticoid supplement. Methods and Results Heart failure was induced by coronary artery ligation for 8 weeks. A total of 32 rats were randomly assigned to 4 groups (n=8/group): sham surgery group, congestive heart failure group, dexamethasone group, and dexamethasone in combination with glucocorticoid receptor antagonist RU486 group. An acute water loading test was administered 6 hours after drug administration. Left ventricular function was measured by a pressure‐volume catheter. Protein expressions were determined by immunohistochemistry and immunoblotting. The pressure‐volume loop analysis showed that dexamethasone improves cardiac function in rats with heart failure. Western blotting confirmed that dexamethasone remarkably reduces the expressions of V2R, aquaporin 2, and aquaporin 3 in the renal‐collecting ducts. As a result of V2R downregulation, the expressions of glucocorticoid regulated kinase 1, apical epithelial sodium channels, and the furosemide‐sensitive Na‐K‐2Cl cotransporter were also downregulated. These favorable effects induced by dexamethasone were mostly abolished by the glucocorticoid receptor inhibitor RU486, indicating that the aforementioned effects are glucocorticoid receptor mediated. Conclusions Glucocorticoids can reverse diluted hyponatremia via inhibiting the vasopressin receptor pathway in rats with heart failure.
Collapse
Affiliation(s)
- Xiaoran Zhu
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China.,Department of Pharmacy Hebei General Hospital Shijiazhuang China
| | - Yaomeng Huang
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China
| | - Shuyu Li
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China.,Department of Cardiovascular Medicine Fengnan District Hospital Tangshan China
| | - Ning Ge
- Regenerative Medicine Institute School of Medicine National University of Ireland Galway Ireland
| | - Tongxin Li
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China
| | - Yu Wang
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China
| | - Kunshen Liu
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China
| | - Chao Liu
- The First Cardiology Division The First Hospital of Hebei Medical University Shijiazhuang China
| |
Collapse
|
29
|
Verschuren EHJ, Castenmiller C, Peters DJM, Arjona FJ, Bindels RJM, Hoenderop JGJ. Sensing of tubular flow and renal electrolyte transport. Nat Rev Nephrol 2020; 16:337-351. [DOI: 10.1038/s41581-020-0259-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
|
30
|
Djamgoz MBA. Hyponatremia and Cancer Progression: Possible Association with Sodium-Transporting Proteins. Bioelectricity 2020; 2:14-20. [PMID: 34471833 DOI: 10.1089/bioe.2019.0035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hyponatremia, the phenomenon of serum sodium level falling below 135 mmol/L, is seen frequently in cancer patients and has been correlated with poor prognosis. Hyponatremia has classically been attributed to the "syndrome of inappropriate antidiuretic hormone secretion," leading to prolonged fluid retention. However, this is unlikely to be the only mechanism. In this study, we advance the hypothesis that upregulation of various sodium-transporting proteins during the cancer process makes a significant contribution to the pathophysiology of cancer-associated hyponatremia. Such sodium-transporting proteins include voltage-gated sodium channels, especially its hypoxia-promoted persistent current, epithelial sodium channels, and transient receptor potential channels. Thus, hyponatremia follows cancer, whereby drop in blood serum level occurs as a result of uptake of sodium from extracellular fluid by cancer cells. Indeed, the sodium content of cancer cells/tissues is higher than normal. In turn, the rise in the intracellular sodium concentration brings about a range of cellular effects, including extracellular acidification that promotes invasiveness and thus leads to poor prognosis. This perspective offers novel therapies for cancer and the associated hyponatremia.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom.,Biotechnology Research Centre, Cyprus International University, Lefkosa, North Cyprus
| |
Collapse
|
31
|
Li Q, Fung E. Multifaceted Functions of Epithelial Na + Channel in Modulating Blood Pressure. Hypertension 2019; 73:273-281. [PMID: 30580685 DOI: 10.1161/hypertensionaha.118.12330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qi Li
- From the Division of Cardiology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (Q.L., E.F.).,Laboratory for Heart Failure and Circulation Research, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Hong Kong SAR (Q.L., E.F.)
| | - Erik Fung
- From the Division of Cardiology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (Q.L., E.F.).,Gerald Choa Cardiac Research Centre, Faculty of Medicine, The Chinese University of Hong Kong (E.F.).,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong (E.F.).,Laboratory for Heart Failure and Circulation Research, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Hong Kong SAR (Q.L., E.F.)
| |
Collapse
|
32
|
Lin SR, Lin SY, Chen CC, Fu YS, Weng CF. Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives. J Clin Med 2019; 8:E2003. [PMID: 31744165 PMCID: PMC6912567 DOI: 10.3390/jcm8112003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
Primary hypertension describes abnormally-high systolic/diastolic blood pressure in a resting condition caused by various genetic or environmental risk factors. Remarkably, severe complications, such as ischemic cardiovascular disease, stroke, and chronic renal disease have led to primary hypertension becoming a huge burden for almost one-third of the total population. Medication is the major regimen for treating primary hypertension; however, recent medications may have adverse effects that attenuate energy levels. Hence, the search for new hypotensive agents from folk or traditional medicine may be fruitful in the discovery and development of new drugs. This review assembles recent findings for natural antihypertensive agents, extracts, or decoctions published in PubMed, and provides insights into the search for new hypotensive compounds based on blood-pressure regulating mechanisms, including the renin-angiotensin-aldosterone system and the sympathetic/adrenergic receptor/calcium channel system.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan; (S.-R.L.); (C.-C.C.)
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan
| | - Shiuan-Yea Lin
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Cheng Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan; (S.-R.L.); (C.-C.C.)
- Camillian Saint Mary’s Hospital Luodong,160 Zhongzheng S. Rd. Luodong, Yilan 26546, Taiwan
| | - Yaw-Syan Fu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Feng Weng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Basic Medical Science, Center for Transitional Medicine, Xiamen Medical College, Xiamen 361023, China
| |
Collapse
|
33
|
“Reversed polarization” of Na/K-ATPase—a sign of inverted transport in the human endolymphatic sac: a super-resolution structured illumination microscopy (SR-SIM) study. Cell Tissue Res 2019; 379:445-457. [DOI: 10.1007/s00441-019-03106-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023]
Abstract
AbstractThe human endolymphatic sac (ES) is believed to regulate inner ear fluid homeostasis and to be associated with Meniere’s disease (MD). We analyzed the ion transport protein sodium/potassium-ATPase (Na/K-ATPase) and its isoforms in the human ES using super-resolution structured illumination microscopy (SR-SIM). Human vestibular aqueducts were collected during trans-labyrinthine vestibular schwannoma surgery after obtaining ethical permission. Antibodies against various isoforms of Na/K-ATPase and additional solute-transporting proteins, believed to be essential for ion and fluid transport, were used for immunohistochemistry. A population of epithelial cells of the human ES strongly expressed Na/K-ATPase α1, β1, and β3 subunit isoforms in either the lateral/basolateral or apical plasma membrane domains. The β1 isoform was expressed in the lateral/basolateral plasma membranes in mostly large cylindrical cells, while β3 and α1 both were expressed with “reversed polarity” in the apical cell membrane in lower epithelial cells. The heterogeneous expression of Na/K-ATPase subunits substantiates earlier notions that the ES is a dynamic structure where epithelial cells show inverted epithelial transport. Dual absorption and secretion processes may regulate and maintain inner ear fluid homeostasis. These findings may shed new light on the etiology of endolymphatic hydrops and MD.
Collapse
|
34
|
Kanbay M, Yilmaz S, Dincer N, Ortiz A, Sag AA, Covic A, Sánchez-Lozada LG, Lanaspa MA, Cherney DZI, Johnson RJ, Afsar B. Antidiuretic Hormone and Serum Osmolarity Physiology and Related Outcomes: What Is Old, What Is New, and What Is Unknown? J Clin Endocrinol Metab 2019; 104:5406-5420. [PMID: 31365096 DOI: 10.1210/jc.2019-01049] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Although the physiology of sodium, water, and arginine vasopressin (AVP), also known as antidiuretic hormone, has long been known, accumulating data suggest that this system operates as a more complex network than previously thought. EVIDENCE ACQUISITION English-language basic science and clinical studies of AVP and osmolarity on the development of kidney and cardiovascular disease and overall outcomes. EVIDENCE SYNTHESIS Apart from osmoreceptors and hypovolemia, AVP secretion is modified by novel factors such as tongue acid-sensing taste receptor cells and brain median preoptic nucleus neurons. Moreover, pharyngeal, esophageal, and/or gastric sensors and gut microbiota modulate AVP secretion. Evidence is accumulating that increased osmolarity, AVP, copeptin, and dehydration are all associated with worse outcomes in chronic disease states such as chronic kidney disease (CKD), diabetes, and heart failure. On the basis of these pathophysiological relationships, an AVP receptor 2 blocker is now licensed for CKD related to polycystic kidney disease. CONCLUSION From a therapeutic perspective, fluid intake may be associated with increased AVP secretion if it is driven by loss of urine concentration capacity or with suppressed AVP if it is driven by voluntary fluid intake. In the current review, we summarize the literature on the relationship between elevated osmolarity, AVP, copeptin, and dehydration with renal and cardiovascular outcomes and underlying classical and novel pathophysiologic pathways. We also review recent unexpected and contrasting findings regarding AVP physiology in an attempt to explain and understand some of these relationships.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sezen Yilmaz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Neris Dincer
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alberto Ortiz
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Adrian Covic
- Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C. I. Parhon" University Hospital, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology, Department of Nephrology, INC Ignacio Chávez, Mexico City, Mexico
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Baris Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| |
Collapse
|
35
|
Edwards A, McDonough AA. Impact of angiotensin II-mediated stimulation of sodium transporters in the nephron assessed by computational modeling. Am J Physiol Renal Physiol 2019; 317:F1656-F1668. [PMID: 31657247 DOI: 10.1152/ajprenal.00335.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Angiotensin II (ANG II) raises blood pressure partly by stimulating tubular Na+ reabsorption. The effects of ANG II on tubular Na+ transporters (i.e., channels, pumps, cotransporters, and exchangers) vary between short-term and long-term exposure. To better understand the physiological impact, we used a computational model of transport along the rat nephron to predict the effects of short- and long-term ANG II-induced transporter activation on Na+ and K+ reabsorption/secretion, and to compare measured and calculated excretion rates. Three days of ANG II infusion at 200 ng·kg-1·min-1 is nonpressor, yet stimulates transporter accumulation. The increase in abundance of Na+/H+ exchanger 3 (NHE3) or activated Na+-K+-2Cl- cotransporter-2 (NKCC2-P) predicted significant reductions in urinary Na+ excretion, yet there was no observed change in urine Na+. The lack of antinatriuresis, despite Na+ transporter accumulation, was supported by Li+ and creatinine clearance measurements, leading to the conclusion that 3-day nonpressor ANG II increases transporter abundance without proportional activation. Fourteen days of ANG II infusion at 400 ng·kg-1·min-1 raises blood pressure and increases Na+ transporter abundance along the distal nephron; proximal tubule and medullary loop transporters are decreased and urine Na+ and volume output are increased, evidence for pressure natriuresis. Simulations indicate that decreases in NHE3 and NKCC2-P contribute significantly to reducing Na+ reabsorption along the nephron and to pressure natriuresis. Our results also suggest that differential regulation of medullary (decrease) and cortical (increase) NKCC2-P is important to preserve K+ while minimizing Na+ retention during ANG II infusion. Lastly, our model indicates that accumulation of active Na+-Cl- cotransporter counteracts epithelial Na+ channel-induced urinary K+ loss.
Collapse
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
36
|
Fructose increases the activity of sodium hydrogen exchanger in renal proximal tubules that is dependent on ketohexokinase. J Nutr Biochem 2019; 71:54-62. [DOI: 10.1016/j.jnutbio.2019.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 11/22/2022]
|
37
|
Liu W, Luque M, Glueckert R, Danckwardt-Lillieström N, Nordström CK, Schrott-Fischer A, Rask-Andersen H. Expression of Na/K-ATPase subunits in the human cochlea: a confocal and super-resolution microscopy study with special reference to auditory nerve excitation and cochlear implantation. Ups J Med Sci 2019; 124:168-179. [PMID: 31460814 PMCID: PMC6758701 DOI: 10.1080/03009734.2019.1653408] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: For the first time the expression of the ion transport protein sodium/potassium-ATPase and its isoforms was analyzed in the human cochlea using light- and confocal microscopy as well as super-resolution structured illumination microscopy. It may increase our understanding of its role in the propagation and processing of action potentials in the human auditory nerve and how electric nerve responses are elicited from auditory prostheses. Material and methods: Archival human cochlear sections were obtained from trans-cochlear surgeries. Antibodies against the Na/K-ATPase β1 isoform together with α1 and α3 were used for immunohistochemistry. An algorithm was applied to assess the expression in various domains. Results: Na/K ATPase β1 subunit was expressed, mostly combined with the α1 isoform. Neurons expressed the β1 subunit combined with α3, while satellite glial cells expressed the α1 isoform without recognized association with β1. Types I and II spiral ganglion neurons and efferent fibers expressed the Na/K-ATPase α3 subunit. Inner hair cells, nerve fibers underneath, and efferent and afferent fibers in the organ of Corti also expressed α1. The highest activity of Na/K-ATPase β1 was at the inner hair cell/nerve junction and spiral prominence. Conclusion: The human auditory nerve displays distinct morphologic features represented in its molecular expression. It was found that electric signals generated via hair cells may not go uninterrupted across the spiral ganglion, but are locally processed. This may be related to particular filtering properties in the human acoustic pathway.
Collapse
Affiliation(s)
- Wei Liu
- Department of Surgical Sciences, Section of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
- Wei Liu
| | - Maria Luque
- Department of Otolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Charlotta Kämpfe Nordström
- Department of Surgical Sciences, Section of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
| | | | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
- CONTACT Helge Rask-Andersen Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85, Uppsala, Sweden
| |
Collapse
|
38
|
Hormonal regulation of Na +-K +-ATPase from the evolutionary perspective. CURRENT TOPICS IN MEMBRANES 2019; 83:315-351. [PMID: 31196608 DOI: 10.1016/bs.ctm.2019.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Na+-K+-ATPase, an α/β heterodimer, is an ancient enzyme that maintains Na+ and K+ gradients, thus preserving cellular ion homeostasis. In multicellular organisms, this basic housekeeping function is integrated to fulfill the needs of specialized organs and preserve whole-body homeostasis. In vertebrates, Na+-K+-ATPase is essential for many fundamental physiological processes, such as nerve conduction, muscle contraction, nutrient absorption, and urine excretion. During vertebrate evolution, three key developments contributed to diversification and integration of Na+-K+-ATPase functions. Generation of novel α- and β-subunits led to formation of multiple Na+-K+-ATPase isoenyzmes with distinct functional characteristics. Development of a complex endocrine system enabled efficient coordination of diverse Na+-K+-ATPase functions. Emergence of FXYDs, small transmembrane proteins that regulate Na+-K+-ATPase, opened new ways to modulate its function. FXYDs are a vertebrate innovation and an important site of hormonal action, suggesting they played an especially prominent role in evolving interaction between Na+-K+-ATPase and the endocrine system in vertebrates.
Collapse
|
39
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Kulkarni NH, Smith RC, Blazer-Yost BL. Loss of inversin decreases transepithelial sodium transport in murine renal cells. Am J Physiol Cell Physiol 2017; 313:C664-C673. [PMID: 28978526 DOI: 10.1152/ajpcell.00359.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Type II nephronophthisis (NPHP2) is an autosomal recessive renal cystic disorder characterized by mutations in the inversin gene. Humans and mice with mutations in inversin have enlarged cystic kidneys that may be due to fluid accumulation resulting from altered ion transport. To address this, transepithelial ion transport was measured in shRNA-mediated inversin-depleted mouse cortical collecting duct (mCCD) cells. Loss of inversin decreased the basal ion flux in mCCD cells compared with controls. Depletion of inversin decreased vasopressin-induced Na+ absorption but did not alter Cl- secretion by mCCD cells. Addition of amiloride, a specific blocker of the epithelial sodium channel (ENaC), abolished basal ion transport in both inversin knockdown and control cells, indicating ENaC involvement. Transcript levels of ENaC β-subunit were reduced in inversin-knockdown cells consistent with decreased ENaC activity. Furthermore, Nedd4l (neural precursor cell expressed, developmentally downregulated 4 like), an upstream negative regulator of ENaC, was evaluated. The relative amount of the phosphorylated, inactive Nedd4l was decreased in inversin-depleted cells consistent with decreased ENaC activity. The protein levels of Sgk1 (serum and glucocorticoid-inducible kinase), which phosphorylates Nedd4l, remained unchanged although the transcript levels were increased in inversin-depleted cells. Interestingly, mRNA and protein levels of Crtc2 (Creb-regulated transcription coactivator) kinase, a positive regulator of Sgk1, were decreased in inversin-depleted cells. Together these results suggest that loss of inversin decreases Na+ transport via ENaC, mediated in part by transcriptional and posttranslational regulation of Crtc2/Sgk1/Nedd4l axis as a contributory mechanism for enlarged kidneys in NPHP2.
Collapse
Affiliation(s)
- Nalini H Kulkarni
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| | - Rosamund C Smith
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| | - Bonnie L Blazer-Yost
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| |
Collapse
|
41
|
Bartoszewski R, Matalon S, Collawn JF. Ion channels of the lung and their role in disease pathogenesis. Am J Physiol Lung Cell Mol Physiol 2017; 313:L859-L872. [PMID: 29025712 PMCID: PMC5792182 DOI: 10.1152/ajplung.00285.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
Maintenance of normal epithelial ion and water transport in the lungs includes providing a thin layer of surface liquid that coats the conducting airways. This airway surface liquid is critical for normal lung function in a number of ways but, perhaps most importantly, is required for normal mucociliary clearance and bacterial removal. Preservation of the appropriate level of hydration, pH, and viscosity for the airway surface liquid requires the proper regulation and function of a battery of different types of ion channels and transporters. Here we discuss how alterations in ion channel/transporter function often lead to lung pathologies.
Collapse
Affiliation(s)
- Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
- Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
- Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
42
|
Rojas M, Díaz P, León P, Gonzalez AA, González M, Barrientos V, Pestov NB, Alzamora R, Michea L. Mineralocorticoids modulate the expression of the β-3 subunit of the Na +, K +-ATPase in the renal collecting duct. Channels (Austin) 2017. [PMID: 28636485 DOI: 10.1080/19336950.2017.1344800] [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: 10/19/2022] Open
Abstract
Renal sodium reabsorption depends on the activity of the Na+,K+-ATPase α/β heterodimer. Four α (α1-4) and 3 β (β1-3) subunit isoforms have been described. It is accepted that renal tubule cells express α1/β1 dimers. Aldosterone stimulates Na+,K+-ATPase activity and may modulate α1/β1 expression. However, some studies suggest the presence of β3 in the kidney. We hypothesized that the β3 isoform of the Na+,K+-ATPase is expressed in tubular cells of the distal nephron, and modulated by mineralocorticoids. We found that β3 is highly expressed in collecting duct of rodents, and that mineralocorticoids decreased the expression of β3. Thus, we describe a novel molecular mechanism of sodium pump modulation that may contribute to the effects of mineralocorticoids on sodium reabsorption.
Collapse
Affiliation(s)
- Macarena Rojas
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Pablo Díaz
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Pablo León
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Alexis A Gonzalez
- b Instituto de Química, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Magdalena González
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Víctor Barrientos
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| | - Nikolay B Pestov
- c Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry , Moscow , Russia.,d Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research , University of Toledo College of Medicine , Toledo , OH , USA
| | - Rodrigo Alzamora
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,e Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Santiago , Chile
| | - Luis Michea
- a Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,f Millennium Institute on Immunology and Immunotherapy , Santiago , Chile
| |
Collapse
|
43
|
McDonough AA, Youn JH. Potassium Homeostasis: The Knowns, the Unknowns, and the Health Benefits. Physiology (Bethesda) 2017; 32:100-111. [PMID: 28202621 PMCID: PMC5337831 DOI: 10.1152/physiol.00022.2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Potassium homeostasis has a very high priority because of its importance for membrane potential. Although extracellular K+ is only 2% of total body K+, our physiology was evolutionarily tuned for a high-K+, low-Na+ diet. We review how multiple systems interface to accomplish fine K+ balance and the consequences for health and disease.
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Jang H Youn
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
44
|
Cuevas CA, Su XT, Wang MX, Terker AS, Lin DH, McCormick JA, Yang CL, Ellison DH, Wang WH. Potassium Sensing by Renal Distal Tubules Requires Kir4.1. J Am Soc Nephrol 2017; 28:1814-1825. [PMID: 28052988 DOI: 10.1681/asn.2016090935] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/23/2016] [Indexed: 11/03/2022] Open
Abstract
The mammalian distal convoluted tubule (DCT) makes an important contribution to potassium homeostasis by modulating NaCl transport. The thiazide-sensitive Na+/Cl- cotransporter (NCC) is activated by low potassium intake and by hypokalemia. Coupled with suppression of aldosterone secretion, activation of NCC helps to retain potassium by increasing electroneutral NaCl reabsorption, therefore reducing Na+/K+ exchange. Yet the mechanisms by which DCT cells sense plasma potassium concentration and transmit the information to the apical membrane are not clear. Here, we tested the hypothesis that the potassium channel Kir4.1 is the potassium sensor of DCT cells. We generated mice in which Kir4.1 could be deleted in the kidney after the mice are fully developed. Deletion of Kir4.1 in these mice led to moderate salt wasting, low BP, and profound potassium wasting. Basolateral membranes of DCT cells were depolarized, nearly devoid of conductive potassium transport, and unresponsive to plasma potassium concentration. Although renal WNK4 abundance increased after Kir4.1 deletion, NCC abundance and function decreased, suggesting that membrane depolarization uncouples WNK kinases from NCC. Together, these results indicate that Kir4.1 mediates potassium sensing by DCT cells and couples this signal to apical transport processes.
Collapse
Affiliation(s)
- Catherina A Cuevas
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Xiao-Tong Su
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Ming-Xiao Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| | - James A McCormick
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon.,Renal Section, Veterans Administration Portland Health Care System, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Departments of Medicine and Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon; .,Renal Section, Veterans Administration Portland Health Care System, Portland, Oregon
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York; and
| |
Collapse
|
45
|
Wilson TE. Renal sympathetic nerve, blood flow, and epithelial transport responses to thermal stress. Auton Neurosci 2016; 204:25-34. [PMID: 28043810 DOI: 10.1016/j.autneu.2016.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/28/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
Thermal stress is a profound sympathetic stress in humans; kidney responses involve altered renal sympathetic nerve activity (RSNA), renal blood flow, and renal epithelial transport. During mild cold stress, RSNA spectral power but not total activity is altered, renal blood flow is maintained or decreased, and epithelial transport is altered consistent with a sympathetic stress coupled with central volume loaded state. Hypothermia decreases RSNA, renal blood flow, and epithelial transport. During mild heat stress, RSNA is increased, renal blood flow is decreased, and epithelial transport is increased consistent with a sympathetic stress coupled with a central volume unloaded state. Hyperthermia extends these directional changes, until heat illness results. Because kidney responses are very difficult to study in humans in vivo, this review describes and qualitatively evaluates an in vivo human skin model of sympathetically regulated epithelial tissue compared to that of the nephron. This model utilizes skin responses to thermal stress, involving 1) increased skin sympathetic nerve activity (SSNA), decreased skin blood flow, and suppressed eccrine epithelial transport during cold stress; and 2) increased SSNA, skin blood flow, and eccrine epithelial transport during heat stress. This model appears to mimic aspects of the renal responses. Investigations of skin responses, which parallel certain renal responses, may aid understanding of epithelial-sympathetic nervous system interactions during cold and heat stress.
Collapse
Affiliation(s)
- Thad E Wilson
- Division of Biomedical Sciences, Marian University College of Osteopathic Medicine, Indianapolis, IN, USA.
| |
Collapse
|