1
|
Otterpohl KL, Busselman BW, Zimmerman JL, Mukherjee M, Evans C, Graber K, Thakkar VP, Johnston JG, Ilyas A, Gumz ML, Eaton DC, Sands JM, Surendran K, Chandrasekar I. Thick Ascending Limb Specific Inactivation of Myh9 and Myh10 Myosin Motors Results in Progressive Kidney Disease and Drives Sex-specific Cellular Adaptation in the Distal Nephron and Collecting Duct. FUNCTION 2025; 6:zqae048. [PMID: 39500539 PMCID: PMC11815580 DOI: 10.1093/function/zqae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
Our previous work established a role for myosin motor proteins MYH9 and MYH10 in trafficking of thick ascending limb (TAL) cargoes uromodulin and Na+-K+-2Cl- cotransporter NKCC2. We have generated a TAL-specific Myh9&10 conditional knockout (Myh9&10 TAL-cKO) mouse model to determine the cell autonomous roles for MYH9&10 in TAL cargo trafficking and to understand the consequence of TAL dysfunction in adult kidney. Myh9&10 TAL-cKO mice develop progressive kidney disease with pathological tubular injury confirmed by histological changes, tubular injury markers, upregulated endoplasmic reticulum (ER) stress/unfolded protein response, and higher blood urea nitrogen and serum creatinine. However, male mice survive twice as long as female mice. We have determined this sexual dimorphism in morbidity is due to adaptation of the distal nephron and collecting duct in response to TAL dysfunction and lower NKCC2 expression. We demonstrate that this triggers a compensatory mechanism involving sex-specific cellular adaptation within the distal nephron and collecting duct to boost sodium reabsorption. While both sexes overcompensate by activating epithelial sodium channel (ENaC) expression in medullary collecting ducts resulting in hypernatremia, this is initially subdued in male Myh9&10 TAL-cKO mice through higher sodium chloride cotransporter (NCC) expression within the distal nephron. Our results indicate that compromised TAL function ultimately results in maladaptation of medullary collecting duct cells which acquire cortical-like properties including ENaC expression. This work further confirms a cell autonomous role for MYH9&10 in maintenance of NKCC2 expression in the TAL and uncover distal nephron and collecting duct adaptive mechanisms which respond to TAL dysfunction.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Malini Mukherjee
- Functional Genomics and Bioinformatics Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Claire Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Kelly Graber
- Histology and Imaging Core, Sanford Research, Sioux Falls, SD 57104, USA
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Jermaine G Johnston
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, SD 57069, USA
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Douglas C Eaton
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Jeff M Sands
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, SD 57103, USA
| |
Collapse
|
2
|
Sakamoto Y, Uezu A, Kikuchi K, Kang J, Fujii E, Moroishi T, Suetsugu S, Nakanishi H. The Nedd4L ubiquitin ligase is activated by FCHO2-generated membrane curvature. EMBO J 2024; 43:5883-5909. [PMID: 39402328 PMCID: PMC11612235 DOI: 10.1038/s44318-024-00268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
The C2-WW-HECT domain ubiquitin ligase Nedd4L regulates membrane sorting during endocytosis through the ubiquitination of cargo molecules such as the epithelial sodium channel (ENaC). Nedd4L is catalytically autoinhibited by an intramolecular interaction between its C2 and HECT domains, but the protein's activation mechanism is poorly understood. Here, we show that Nedd4L activation is linked to membrane shape by FCHO2, a Bin-Amphiphysin-Rsv (BAR) domain protein that regulates endocytosis. FCHO2 was required for the Nedd4L-mediated ubiquitination and endocytosis of ENaC, with Nedd4L co-localizing with FCHO2 at clathrin-coated pits. In cells, Nedd4L was specifically recruited to, and activated by, the FCHO2 BAR domain. Furthermore, we reconstituted FCHO2-induced recruitment and activation of Nedd4L in vitro. Both the recruitment and activation were mediated by membrane curvature rather than protein-protein interactions. The Nedd4L C2 domain recognized a specific degree of membrane curvature that was generated by the FCHO2 BAR domain, with this curvature directly activating Nedd4L by relieving its autoinhibition. Thus, we show for the first time a specific function (i.e., recruitment and activation of an enzyme regulating cargo sorting) of membrane curvature by a BAR domain protein.
Collapse
Affiliation(s)
- Yasuhisa Sakamoto
- Department of Molecular Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjyo, Kumamoto, 860-8556, Japan
| | - Akiyoshi Uezu
- Department of Molecular Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjyo, Kumamoto, 860-8556, Japan
| | - Koji Kikuchi
- Department of Molecular Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjyo, Kumamoto, 860-8556, Japan
| | - Jangmi Kang
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23 Morikita-machi, Kobe, 658-0001, Japan
| | - Eiko Fujii
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23 Morikita-machi, Kobe, 658-0001, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjyo, Kumamoto, 860-8556, Japan
| | - Shiro Suetsugu
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, 630-0192, Japan
| | - Hiroyuki Nakanishi
- Department of Molecular Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjyo, Kumamoto, 860-8556, Japan.
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23 Morikita-machi, Kobe, 658-0001, Japan.
| |
Collapse
|
3
|
Ho QV, Young MJ. Mineralocorticoid receptors, macrophages and new mechanisms for cardiovascular disease. Mol Cell Endocrinol 2024; 593:112340. [PMID: 39134137 DOI: 10.1016/j.mce.2024.112340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Quoc Viet Ho
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia; Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Kuralay A, McDonough MC, Resch JM. Control of sodium appetite by hindbrain aldosterone-sensitive neurons. Mol Cell Endocrinol 2024; 592:112323. [PMID: 38936597 PMCID: PMC11381173 DOI: 10.1016/j.mce.2024.112323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024]
Abstract
Mineralocorticoids play a key role in hydromineral balance by regulating sodium retention and potassium wasting. Through favoring sodium, mineralocorticoids can cause hypertension from fluid overload under conditions of hyperaldosteronism, such as aldosterone-secreting tumors. An often-overlooked mechanism by which aldosterone functions to increase sodium is through stimulation of salt appetite. To drive sodium intake, aldosterone targets neurons in the hindbrain which uniquely express 11β-hydroxysteroid dehydrogenase type 2 (HSD2). This enzyme is a necessary precondition for aldosterone-sensing cells as it metabolizes glucocorticoids - preventing their activation of the mineralocorticoid receptor. In this review, we will consider the role of hindbrain HSD2 neurons in regulating sodium appetite by discussing HSD2 expression in the brain, regulation of hindbrain HSD2 neuron activity, and the circuitry mediating the effects of these aldosterone-sensitive neurons. Reducing the activity of hindbrain HSD2 neurons may be a viable strategy to reduce sodium intake and cardiovascular risk, particularly for conditions of hyperaldosteronism.
Collapse
Affiliation(s)
- Ahmet Kuralay
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Miriam C McDonough
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
5
|
Sasidharan A, Grosche A, Xu X, Kinane TB, Angoli D, Vidyasagar S. Select amino acids recover cytokine-altered ENaC function in human bronchial epithelial cells. PLoS One 2024; 19:e0307809. [PMID: 39052685 PMCID: PMC11271875 DOI: 10.1371/journal.pone.0307809] [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: 10/01/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
The airway epithelium plays a pivotal role in regulating mucosal immunity and inflammation. Epithelial barrier function, homeostasis of luminal fluid, and mucociliary clearance are major components of mucosal defense mechanisms. The epithelial sodium channel (ENaC) is one of the key players in controlling airway fluid volume and composition, and characteristic cytokines cause ENaC and barrier dysfunctions following pulmonary infections or allergic reactions. Given the limited understanding of the requisite duration and magnitude of cytokines to affect ENaC and barrier function, available treatment options for restoring normal ENaC activity are limited. Previous studies have demonstrated that distinct amino acids can modulate epithelial ion channel activities and barrier function in intestines and airways. Here, we have investigated the time- and concentration-dependent effect of representative cytokines for Th1- (IFN-γ and TNF-α), Th2- (IL-4 and IL-13), and Treg-mediated (TGF-β1) immune responses on ENaC activity and barrier function in human bronchial epithelial cells. When cells were exposed to Th1 and Treg cytokines, ENaC activity decreased gradually while barrier function remained largely unaffected. In contrast, Th2 cytokines had an immediate and profound inhibitory effect on ENaC activity that was subsequently followed by epithelial barrier disruption. These functional changes were associated with decreased membrane protein expression of α-, β-, and γ-ENaC, and decreased mRNA levels of β- and γ-ENaC. A proprietary blend of amino acids was developed based on their ability to prevent Th2 cytokine-induced ENaC dysfunction. Exposure to the select amino acids reversed the inhibitory effect of IL-13 on ENaC activity by increasing mRNA levels of β- and γ-ENaC, and protein expression of γ-ENaC. This study indicates the beneficial effect of select amino acids on ENaC activity in an in vitro setting of Th2-mediated inflammation suggesting these amino acids as a novel therapeutic approach for correcting this condition.
Collapse
Affiliation(s)
- Anusree Sasidharan
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Astrid Grosche
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Xiaodong Xu
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - T. Bernard Kinane
- Pediatric Pulmonary Division, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Damiano Angoli
- Pediatric Pulmonary Division, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
6
|
Aufy M, Hussein AM, Stojanovic T, Studenik CR, Kotob MH. Proteolytic Activation of the Epithelial Sodium Channel (ENaC): Its Mechanisms and Implications. Int J Mol Sci 2023; 24:17563. [PMID: 38139392 PMCID: PMC10743461 DOI: 10.3390/ijms242417563] [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: 11/20/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Epithelial sodium channel (ENaC) are integral to maintaining salt and water homeostasis in various biological tissues, including the kidney, lung, and colon. They enable the selective reabsorption of sodium ions, which is a process critical for controlling blood pressure, electrolyte balance, and overall fluid volume. ENaC activity is finely controlled through proteolytic activation, a process wherein specific enzymes, or proteases, cleave ENaC subunits, resulting in channel activation and increased sodium reabsorption. This regulatory mechanism plays a pivotal role in adapting sodium transport to different physiological conditions. In this review article, we provide an in-depth exploration of the role of proteolytic activation in regulating ENaC activity. We elucidate the involvement of various proteases, including furin-like convertases, cysteine, and serine proteases, and detail the precise cleavage sites and regulatory mechanisms underlying ENaC activation by these proteases. We also discuss the physiological implications of proteolytic ENaC activation, focusing on its involvement in blood pressure regulation, pulmonary function, and intestinal sodium absorption. Understanding the mechanisms and consequences of ENaC proteolytic activation provides valuable insights into the pathophysiology of various diseases, including hypertension, pulmonary disorders, and various gastrointestinal conditions. Moreover, we discuss the potential therapeutic avenues that emerge from understanding these mechanisms, offering new possibilities for managing diseases associated with ENaC dysfunction. In summary, this review provides a comprehensive discussion of the intricate interplay between proteases and ENaC, emphasizing the significance of proteolytic activation in maintaining sodium and fluid balance in both health and disease.
Collapse
Affiliation(s)
- Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Ahmed M. Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Mohamed H. Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
7
|
Charoensri S, Auchus RJ. Therapeutic management of congenital forms of endocrine hypertension. Eur J Endocrinol 2023; 189:R11-R22. [PMID: 37847213 DOI: 10.1093/ejendo/lvad140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
Congenital forms of endocrine hypertension are rare and potentially life-threatening disorders, primarily caused by genetic defects affecting adrenal steroid synthesis and activation pathways. These conditions exhibit diverse clinical manifestations, which can be distinguished by their unique molecular mechanisms and steroid profiles. Timely diagnosis and customized management approach are crucial to mitigate unfavorable outcomes associated with uncontrolled hypertension and other related conditions. Treatment options for these disorders depend on the distinct underlying pathophysiology, which involves specific pharmacological therapies or surgical adrenalectomy in some instances. This review article summarizes the current state of knowledge on the therapeutic management of congenital forms of endocrine hypertension, focusing on familial hyperaldosteronism (FH), congenital adrenal hyperplasia, apparent mineralocorticoid excess, and Liddle syndrome. We provide an overview of the genetic and molecular pathogenesis underlying each disorder, describe the clinical features, and discuss the various therapeutic approaches available and their risk of adverse effects, aiming to improve outcomes in patients with these rare and complex conditions.
Collapse
Affiliation(s)
- Suranut Charoensri
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States
- Endocrinology & Metabolism Section, Medicine Service, LTC Charles S. Kettles VA Medical Center, Ann Arbor, MI 48104, United States
| |
Collapse
|
8
|
Wang S, Ma J, Li X, Xian X, Tan G, Cai H, Yang B, Zhang A, Guo J, Gu G, Meng Z, Fu B. EGR-1 Contributes to Pulmonary Edema by Regulating the Epithelial Sodium Channel in Lipopolysaccharide-Induced Acute Lung Injury. Immunol Invest 2023; 52:925-939. [PMID: 37732637 DOI: 10.1080/08820139.2023.2256778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Acute lung injury (ALI) is a common lung disease with increasing morbidity and mortality rates due to the lack of specific drugs. Impaired alveolar fluid clearance (AFC) is a primary pathological feature of ALI. Epithelial sodium channel (ENaC) is a primary determinant in regulating the transport of Na+ and the clearance of alveolar edema fluid. Therefore, ENaC is an important target for the development of drugs for ALI therapy. However, the role of ENaC in the progression of ALI remains unclear. Inhibition of early growth response factor (EGR-1) expression has been reported to induce a protective effect on ALI; therefore, we evaluated whether EGR-1 participates in the progression of ALI by regulating ENaC-α in alveolar epithelium. We investigated the potential mechanism of EGR-1-mediated regulation of ENaC in ALI. We investigated whether EGR-1 aggravates the pulmonary edema response in ALI by regulating ENaC. ALI mouse models were established by intrabronchial injection of lipopolysaccharides (LPS). Lentiviruses with EGR-1 knockdown were transfected into LPS-stimulated A549 cells. We found that EGR-1 expression was upregulated in the lung tissues of ALI mice and in LPS-induced A549 cells, and was negatively correlated with ENaC-α expression. Knockdown of EGR-1 increased ENaC-α expression and relieved cellular edema in ALI. Moreover, EGR-1 regulated ENaC-α expression at the transcriptional level, and correspondingly promoted pulmonary edema and aggravated ALI symptoms. In conclusion, our study demonstrated that EGR-1 could promote pulmonary edema by downregulating ENaC-α at the transcriptional level in ALI. Our study provides a new potential therapeutic strategy for treatment of ALI.
Collapse
Affiliation(s)
- Song Wang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jing Ma
- Office of Academic Research, Liaocheng People's Hospital, Liaocheng, China
| | - Xin Li
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Xinmiao Xian
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guikun Tan
- Pharmacy Department, Liaocheng Woman and Child Health Care Hospital, Liaocheng, China
| | - Hongwei Cai
- Department of Critical Care Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Bingwu Yang
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Anqi Zhang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Jianran Guo
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Guohao Gu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Zhen Meng
- Biomedical Laboratory, Medical School of Liaocheng University, Liaocheng, China
| | - Bo Fu
- Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
9
|
Ismail N, Myint K, Khaing SL, Giribabu N, Salleh N. Cervical fluid pH, electrolytes and osmolarity changes and expression of ion transporters (ENaC, CFTR and AQP) in cervix of women with primary unexplained infertility. Mol Biol Rep 2023; 50:6729-6737. [PMID: 37382776 DOI: 10.1007/s11033-023-08555-6] [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: 10/18/2022] [Accepted: 05/30/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Unexplained infertility could arise from a defect in the cervix. However, the contribution of abnormal cervical fluid microenvironment to this problem still needs to be identified. Therefore, this study identifies the changes in the cervical fluid microenvironment, i.e., pH, electrolytes and osmolarity as well as expression of ion transporters in the cervix including ENaC, CFTR and AQP in fertile women and in women suffering from primary unexplained infertility. METHODS Fertile women and women with unexplained infertility but having regular 28-day menstrual cycles were chosen in this study, Day-22 serum progesterone levels were determined. In the meantime, serum FSH and LH levels were determined on day 2 while, cervical flushing was performed at day 14 to analyse changes in the cervical fluid pH, osmolarity, Na+ and Cl- levels. Meanwhile, cells retrieved from cervical fluid were subjected to mRNA expression and protein distribution analysis for CFTR, AQP and ENaC by qPCR and immunofluorescence, respectively. RESULTS No significant changes in serum progesterone, FSH and LH levels were observed between the two groups. However, cervical fluid pH, osmolarity, Na+ and Cl- levels were significantly lower in primary unexplained infertile group when compared to fertile group. Expression of CFTR and AQP (AQP 1, AQP 2, AQP 5 and AQP 7) in endocervical cells was lower and expression of β-ENaC was higher in primary unexplained infertile women (p < 0.05 when compared to fertile group). CONCLUSIONS Alterations in the cervical fluid microenvironment linked to the defective ion transporter expression in the cervix might contribute towards the unfavourable condition that accounts for unexplained infertility in women.
Collapse
Affiliation(s)
- Nurain Ismail
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kyaimon Myint
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Si Lay Khaing
- Department of Obstetrics & Gynaecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
- Medical Education Department, University of Medicine, Yangon, Myanmar
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
10
|
Zhang YJ, Yang C, Wang W, Harafuji N, Stasiak P, Bell PD, Caldovic L, Sztul E, Guay‐Woodford LM, Bebok Z. Cystin is required for maintaining fibrocystin (FPC) levels and safeguarding proteome integrity in mouse renal epithelial cells: A mechanistic connection between the kidney defects in cpk mice and human ARPKD. FASEB J 2023; 37:e23008. [PMID: 37318790 PMCID: PMC10929748 DOI: 10.1096/fj.202300100r] [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: 01/20/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is caused primarily by mutations in PKHD1, encoding fibrocystin (FPC), but Pkhd1 mutant mice failed to reproduce the human phenotype. In contrast, the renal lesion in congenital polycystic kidney (cpk) mice, with a mutation in Cys1 and cystin protein loss, closely phenocopies ARPKD. Although the nonhomologous mutation diminished the translational relevance of the cpk model, recent identification of patients with CYS1 mutations and ARPKD prompted the investigations described herein. We examined cystin and FPC expression in mouse models (cpk, rescued-cpk (r-cpk), Pkhd1 mutants) and mouse cortical collecting duct (CCD) cell lines (wild type (wt), cpk). We found that cystin deficiency caused FPC loss in both cpk kidneys and CCD cells. FPC levels increased in r-cpk kidneys and siRNA of Cys1 in wt cells reduced FPC. However, FPC deficiency in Pkhd1 mutants did not affect cystin levels. Cystin deficiency and associated FPC loss impacted the architecture of the primary cilium, but not ciliogenesis. No reduction in Pkhd1 mRNA levels in cpk kidneys and CCD cells suggested posttranslational FPC loss. Studies of cellular protein degradation systems suggested selective autophagy as a mechanism. In support of the previously described function of FPC in E3 ubiquitin ligase complexes, we demonstrated reduced polyubiquitination and elevated levels of functional epithelial sodium channel in cpk cells. Therefore, our studies expand the function of cystin in mice to include inhibition of Myc expression via interaction with necdin and maintenance of FPC as functional component of the NEDD4 E3 ligase complexes. Loss of FPC from E3 ligases may alter the cellular proteome, contributing to cystogenesis through multiple, yet to be defined, mechanisms.
Collapse
Affiliation(s)
- Yiming J. Zhang
- Department of Cell Developmental and Integrative Biology (CDIB)University of Alabama at Birmingham, School of MedicineBirminghamAlabamaUSA
| | - Chaozhe Yang
- Center for Translational ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Wei Wang
- Cystic Fibrosis Research CenterUniversity of Alabama at Birmingham, School of MedicineBirminghamAlabamaUSA
| | - Naoe Harafuji
- Center for Translational ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Piotr Stasiak
- Department of Cell Developmental and Integrative Biology (CDIB)University of Alabama at Birmingham, School of MedicineBirminghamAlabamaUSA
| | - P. Darwin Bell
- Department of Medicine, Division of NephrologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Ljubica Caldovic
- Center for Translational ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Elizabeth Sztul
- Department of Cell Developmental and Integrative Biology (CDIB)University of Alabama at Birmingham, School of MedicineBirminghamAlabamaUSA
| | - Lisa M. Guay‐Woodford
- Center for Translational ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Zsuzsanna Bebok
- Department of Cell Developmental and Integrative Biology (CDIB)University of Alabama at Birmingham, School of MedicineBirminghamAlabamaUSA
| |
Collapse
|
11
|
Li Y, Martin TE, Hancock JM, Li R, Viswanathan S, Lydon JP, Zheng Y, Ye X. Visualization of preimplantation uterine fluid absorption in mice using Alexa Fluor™ 488 Hydrazide†. Biol Reprod 2023; 108:204-217. [PMID: 36308434 PMCID: PMC9930399 DOI: 10.1093/biolre/ioac198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/06/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Uterine fluid plays important roles in supporting early pregnancy events and its timely absorption is critical for embryo implantation. In mice, its volume is maximum on day 0.5 post-coitum (D0.5) and approaches minimum upon embryo attachment ~D4.0. Its secretion and absorption in ovariectomized rodents were shown to be promoted by estrogen and progesterone (P4), respectively. The temporal mechanisms in preimplantation uterine fluid absorption remain to be elucidated. We have established an approach using intraluminally injected Alexa Fluor™ 488 Hydrazide (AH) in preimplantation control (RhoAf/f) and P4-deficient RhoAf/fPgrCre/+ mice. In control mice, bulk entry (seen as smeared cellular staining) via uterine luminal epithelium (LE) decreases from D0.5 to D3.5. In P4-deficient RhoAf/fPgrCre/+ mice, bulk entry on D0.5 and D3.5 is impaired. Exogenous P4 treatment on D1.5 and D2.5 increases bulk entry in D3.5 P4-deficient RhoAf/fPgrCre/+ LE, while progesterone receptor (PR) antagonist RU486 treatment on D1.5 and D2.5 diminishes bulk entry in D3.5 control LE. The abundance of autofluorescent apical fine dots, presumptively endocytic vesicles to reflect endocytosis, in the LE cells is generally increased from D0.5 to D3.5 but its regulation by exogenous P4 or RU486 is not obvious under our experimental setting. In the glandular epithelium (GE), bulk entry is rarely observed and green cellular dots do not show any consistent differences among all the investigated conditions. This study demonstrates the dominant role of LE but not GE, the temporal mechanisms of bulk entry and endocytosis in the LE, and the inhibitory effects of P4-deficiency and RU486 on bulk entry in the LE in preimplantation uterine fluid absorption.
Collapse
Affiliation(s)
- Yuehuan Li
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Taylor Elijah Martin
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Jonathan Matthew Hancock
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Rong Li
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Suvitha Viswanathan
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Research Foundation, Cincinnati, Ohio, USA
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
12
|
Scindia YM, Gholam MF, Waleed A, Liu LP, Chacko KM, Desai D, Lopez JP, Malik Z, Schramm WC, Morales AG, Carson-Marino M, Alli AA. Metformin Alleviates Diabetes-Associated Hypertension by Attenuating the Renal Epithelial Sodium Channel. Biomedicines 2023; 11:305. [PMID: 36830842 PMCID: PMC9953274 DOI: 10.3390/biomedicines11020305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Diabetic nephropathy is the primary cause of morbidity in type 2 diabetes mellitus (T2DM) patients. New data indicate that hypertension, a common comorbidity in T2DM, can worsen outcomes of diabetic nephropathy. While metformin is a commonly prescribed drug for treating type 2 diabetes, its blood pressure regulating ability is not well documented. The aim of this study was to investigate the effect of metformin on normalizing blood pressure in salt-loaded hypertensive diabetic db/db mice. Sixteen-week-old male and female diabetic db/db mice were individually placed in metabolic cages and then randomized to a control vehicle (saline) or metformin treatment group. We evaluated the blood pressure reducing ability of metformin in salt-induced hypertension and progression of nephropathy in db/db mice. We observed that metformin- normalized systolic blood pressure in hypertensive diabetic mice. Mechanistically, metformin treatment reduced renal cathepsin B expression. Low cathepsin B expression was associated with reduced expression and activity of the epithelial sodium channel (ENaC), sodium retention, and thus control of hypertension. In addition, we identified that urinary extracellular vesicles (EVs) from the diabetic mice are enriched in cathepsin B. Compared to treatment with urinary EVs of vehicle-treated hypertensive diabetic mice, the amiloride-sensitive transepithelial current was significantly attenuated upon exposure of renal collecting duct cells to urinary EVs isolated from metformin-treated db/db mice or cathepsin B knockout mice. Collectively, our study identifies a novel blood pressure reducing role of metformin in diabetic nephropathy by regulating the cathepsin B-ENaC axis.
Collapse
Affiliation(s)
- Yogesh M. Scindia
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pathology, University of Florida, Gainesville, FL 32610, USA
| | - Mohammed F. Gholam
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
| | - Alina Waleed
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lauren P. Liu
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kevin M. Chacko
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dhruv Desai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Juliana Pena Lopez
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeeshan Malik
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Whitney C. Schramm
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Angelica G. Morales
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Morgan Carson-Marino
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Abdel A. Alli
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
13
|
Jang H, Park Y, Jang J. Serum and glucocorticoid-regulated kinase 1: Structure, biological functions, and its inhibitors. Front Pharmacol 2022; 13:1036844. [PMID: 36457711 PMCID: PMC9706101 DOI: 10.3389/fphar.2022.1036844] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase belonging to the protein kinase A, G, and C (AGC) family. Upon initiation of the phosphoinositide 3-kinase (PI3K) signaling pathway, mammalian target of rapamycin complex 2 (mTORC2) and phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylate the hydrophobic motif and kinase domain of SGK1, respectively, inducing SGK1 activation. SGK1 modulates essential cellular processes such as proliferation, survival, and apoptosis. Hence, dysregulated SGK1 expression can result in multiple diseases, including hypertension, cancer, autoimmunity, and neurodegenerative disorders. This review provides a current understanding of SGK1, particularly in sodium transport, cancer progression, and autoimmunity. In addition, we summarize the developmental status of SGK1 inhibitors, their structures, and respective potencies evaluated in pre-clinical experimental settings. Collectively, this review highlights the significance of SGK1 and proposes SGK1 inhibitors as potential drugs for treatment of clinically relevant diseases.
Collapse
Affiliation(s)
- Hyunsoo Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Youngjun Park
- Laboratory of Immune and Inflammatory Disease, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, South Korea
| | - Jaebong Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| |
Collapse
|
14
|
Kaulich E, Grundy LJ, Schafer WR, Walker DS. The diverse functions of the DEG/ENaC family: linking genetic and physiological insights. J Physiol 2022; 601:1521-1542. [PMID: 36314992 PMCID: PMC10148893 DOI: 10.1113/jp283335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
The DEG/ENaC family of ion channels was defined based on the sequence similarity between degenerins (DEG) from the nematode Caenorhabditis elegans and subunits of the mammalian epithelial sodium channel (ENaC), and also includes a diverse array of non-voltage-gated cation channels from across animal phyla, including the mammalian acid-sensing ion channels (ASICs) and Drosophila pickpockets. ENaCs and ASICs have wide ranging medical importance; for example, ENaCs play an important role in respiratory and renal function, and ASICs in ischaemia and inflammatory pain, as well as being implicated in memory and learning. Electrophysiological approaches, both in vitro and in vivo, have played an essential role in establishing the physiological properties of this diverse family, identifying an array of modulators and implicating them in an extensive range of cellular functions, including mechanosensation, acid sensation and synaptic modulation. Likewise, genetic studies in both invertebrates and vertebrates have played an important role in linking our understanding of channel properties to function at the cellular and whole animal/behavioural level. Drawing together genetic and physiological evidence is essential to furthering our understanding of the precise cellular roles of DEG/ENaC channels, with the diversity among family members allowing comparative physiological studies to dissect the molecular basis of these diverse functions.
Collapse
Affiliation(s)
- Eva Kaulich
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura J Grundy
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK.,Department of Biology, KU Leuven, Leuven, Belgium
| | - Denise S Walker
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
15
|
Blaconà G, Raso R, Castellani S, Pierandrei S, Del Porto P, Ferraguti G, Ascenzioni F, Conese M, Lucarelli M. Downregulation of epithelial sodium channel (ENaC) activity in cystic fibrosis cells by epigenetic targeting. Cell Mol Life Sci 2022; 79:257. [PMID: 35462606 PMCID: PMC9035428 DOI: 10.1007/s00018-022-04190-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 12/31/2022]
Abstract
The pathogenic mechanism of cystic fibrosis (CF) includes the functional interaction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein with the epithelial sodium channel (ENaC). The reduction of ENaC activity may constitute a therapeutic option for CF. This hypothesis was evaluated using drugs that target the protease-dependent activation of the ENaC channel and the transcriptional activity of its coding genes. To this aim we used: camostat, a protease inhibitor; S-adenosyl methionine (SAM), showed to induce DNA hypermethylation; curcumin, known to produce chromatin condensation. SAM and camostat are drugs already clinically used in other pathologies, while curcumin is a common dietary compound. The experimental systems used were CF and non-CF immortalized human bronchial epithelial cell lines as well as human bronchial primary epithelial cells. ENaC activity and SCNN1A, SCNN1B and SCNN1G gene expression were analyzed, in addition to SCNN1B promoter methylation. In both immortalized and primary cells, the inhibition of extracellular peptidases and the epigenetic manipulations reduced ENaC activity. Notably, the reduction in primary cells was much more effective. The SCNN1B appeared to be the best target to reduce ENaC activity, in respect to SCNN1A and SCNN1G. Indeed, SAM treatment resulted to be effective in inducing hypermethylation of SCNN1B gene promoter and in lowering its expression. Importantly, CFTR expression was unaffected, or even upregulated, after treatments. These results open the possibility of CF patients’ treatment by epigenetic targeting.
Collapse
Affiliation(s)
- Giovanna Blaconà
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberto Raso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefano Castellani
- Department of Biomedical Sciences and Human Oncology, University of Bari, Bari, Italy
| | - Silvia Pierandrei
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Paola Del Porto
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy. .,Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
16
|
Tomilin VN, Pyrshev K, Stavniichuk A, Hassanzadeh Khayyat N, Ren G, Zaika O, Khedr S, Staruschenko A, Mei FC, Cheng X, Pochynyuk O. Epac1-/- and Epac2-/- mice exhibit deficient epithelial Na+ channel regulation and impaired urinary Na+ conservation. JCI Insight 2022; 7:e145653. [PMID: 34914636 PMCID: PMC8855822 DOI: 10.1172/jci.insight.145653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/15/2021] [Indexed: 12/03/2022] Open
Abstract
Exchange proteins directly activated by cAMP (Epacs) are abundantly expressed in the renal tubules. We used genetic and pharmacological tools in combination with balance, electrophysiological, and biochemical approaches to examine the role of Epac1 and Epac2 in renal sodium handling. We demonstrate that Epac1-/- and Epac2-/- mice exhibit a delayed anti-natriuresis to dietary sodium restriction despite augmented aldosterone levels. This was associated with a significantly lower response to the epithelial Na+ channel (ENaC) blocker amiloride, reduced ENaC activity in split-opened collecting ducts, and defective posttranslational processing of α and γENaC subunits in the KO mice fed with a Na+-deficient diet. Concomitant deletion of both isoforms led to a marginally greater natriuresis but further increased aldosterone levels. Epac2 blocker ESI-05 and Epac1&2 blocker ESI-09 decreased ENaC activity in Epac WT mice kept on the Na+-deficient diet but not on the regular diet. ESI-09 injections led to natriuresis in Epac WT mice on the Na+-deficient diet, which was caused by ENaC inhibition. In summary, our results demonstrate similar but nonredundant actions of Epac1 and Epac2 in stimulation of ENaC activity during variations in dietary salt intake. We speculate that inhibition of Epac signaling could be instrumental in treatment of hypertensive states associated with ENaC overactivation.
Collapse
Affiliation(s)
- Viktor N. Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kyrylo Pyrshev
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anna Stavniichuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Naghmeh Hassanzadeh Khayyat
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Guohui Ren
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Physiology, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Fang C. Mei
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
17
|
Palma AG, Kotsias BA. The Effect of Dynasore Upon the Negative Interaction Between ENaC and CFTR Channels in Xenopus laevis Oocytes. J Membr Biol 2022; 255:61-69. [DOI: 10.1007/s00232-021-00212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022]
|
18
|
Orr-Burks N, Murray J, Todd KV, Bakre A, Tripp RA. Drug repositioning of Clopidogrel or Triamterene to inhibit influenza virus replication in vitro. PLoS One 2021; 16:e0259129. [PMID: 34714852 PMCID: PMC8555795 DOI: 10.1371/journal.pone.0259129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022] Open
Abstract
Influenza viruses cause respiratory tract infections and substantial health concerns. Infection may result in mild to severe respiratory disease associated with morbidity and some mortality. Several anti-influenza drugs are available, but these agents target viral components and are susceptible to drug resistance. There is a need for new antiviral drug strategies that include repurposing of clinically approved drugs. Drugs that target cellular machinery necessary for influenza virus replication can provide a means for inhibiting influenza virus replication. We used RNA interference screening to identify key host cell genes required for influenza replication, and then FDA-approved drugs that could be repurposed for targeting host genes. We examined the effects of Clopidogrel and Triamterene to inhibit A/WSN/33 (EC50 5.84 uM and 31.48 uM, respectively), A/CA/04/09 (EC50 6.432 uM and 3.32 uM, respectively), and B/Yamagata/16/1988 (EC50 0.28 uM and 0.11 uM, respectively) replication. Clopidogrel and Triamterene provide a druggable approach to influenza treatment across multiple strains and subtypes.
Collapse
Affiliation(s)
- Nichole Orr-Burks
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Kyle V. Todd
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Abhijeet Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
19
|
Pandey RK, Pandey RK, Shukla SS, Pandey P. A review on corona virus and treatment approaches with Allium sativam. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:159. [PMID: 34395639 PMCID: PMC8353433 DOI: 10.1186/s43094-021-00310-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/04/2021] [Indexed: 11/13/2022] Open
Abstract
Background Recently reported cases of Covid-19 globally remind us that new diseases are coming while we are unable to provide the treatment for the same. The entire world is facing this viral attack; deaths are increasing day by day as well as infected patients too. Today, in the period of this disease, can we go to the shelter of our
traditional medicines? Main body In this article, we have taken medicines related to corona and conceptualized their mechanism, which gave us a chance to understand Garlic's mechanism of action, how Garlic can be a weapon in the lane with this disease. This article also tells how we can treat new diseases with our traditional herbs if no modern medicine has been discovered yet. Conclusion The present review is based on the structure of the virus and the targeted site for the drug discovery process with important constituents of Allium sativam. The review work also explains the allicin chemical constituent of Allium sativam which has targeted therapeutic sites related to Covid-19.
Collapse
Affiliation(s)
- Rupesh Kumar Pandey
- Department of Pharmacology KSCP, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh India
| | | | | | - Priyanka Pandey
- Swami Vivekanand College of Pharmacy, Indore, Madhya Pradesh India
| |
Collapse
|
20
|
Zhang J, Yuan HK, Chen S, Zhang ZR. Detrimental or beneficial: Role of endothelial ENaC in vascular function. J Cell Physiol 2021; 237:29-48. [PMID: 34279047 DOI: 10.1002/jcp.30505] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/19/2022]
Abstract
In the past, it was believed that the expression of the epithelial sodium channel (ENaC) was restricted to epithelial tissues, such as the distal nephron, airway, sweat glands, and colon, where it is critical for sodium homeostasis. Over the past two decades, this paradigm has shifted due to the finding that ENaC is also expressed in various nonepithelial tissues, notably in vascular endothelial cells. In this review, the recent findings of the expression, regulation, and function of the endothelial ENaC (EnNaC) are discussed. The expression of EnNaC subunits is reported in a variety of endothelial cell lines and vasculatures, but this is controversial across different species and vessels and is not a universal finding in all vascular beds. The expression density of EnNaC is very faint compared to ENaC in the epithelium. To date, little is known about the regulatory mechanism of EnNaC. Through it can be regulated by aldosterone, the detailed downstream signaling remains elusive. EnNaC responds to increased extracellular sodium with the feedforward activation mechanism, which is quite different from the Na+ self-inhibition mechanism of ENaC. Functionally, EnNaC was shown to be a determinant of cellular mechanics and vascular tone as it can sense shear stress, and its activation or insertion into plasma membrane causes endothelial stiffness and reduced nitric oxide production. However, in some blood vessels, EnNaC is essential for maintaining the integrity of endothelial barrier function. In this context, we discuss the possible reasons for the distinct role of EnNaC in vasculatures.
Collapse
Affiliation(s)
- Jun Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Kai Yuan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Chen
- Department of Biopharmaceutical Sciences, School of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Zhi-Ren Zhang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University & Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| |
Collapse
|
21
|
Ejaz S, Ali A, Azim K, Mahmood A, Khan AI, Almazyad TA, Bilal B. Association between preeclampsia and prostasin polymorphism in Pakistani females. Saudi Med J 2021; 41:1234-1240. [PMID: 33130844 PMCID: PMC7804221 DOI: 10.15537/smj.2020.11.25497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objectives: To investigate the relationship between a prostasin gene variations and the development of preeclampsia in a Pakistani female population. Methods: This was a case-control study carried out at University of Karachi, Karachi, Pakistan between May 2018 and 2019. A single nucleotide polymorphism (SNP) at rs12597511 locus was examined with polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP) analyses in 76 preeclamptic and 74 normotensive expecting mothers. Results: We observed significantly increased risk of preeclampsia associated with the CC genotype of rs12597511 polymorphism as compared to TT (p<0.001, OR=8.08, 95% CI:1.28-31.19) and TT/TC (p<0.001, OR=14.66 and 95% CI: 3.31-65.07) genotypes carriers. Calculation of the allelic distribution revealed a higher frequency of the T allele (82%) among controls; however, the C allele was more prevalent in the preeclamptic group (36%) significantly. Conclusion: The significantly higher C allele frequency in the prostasin gene at the rs12597511 locus in the preeclamptic group indicates that the distribution of the C allele of the prostasin gene is a potential risk factor contributing to the development of preeclampsia.
Collapse
Affiliation(s)
- Saima Ejaz
- Department of Physiology, King Saud Bin Abdulaziz University of Health Sciences, Riyadh, Kingdom of Saudi Arabia. E-mail.
| | | | | | | | | | | | | |
Collapse
|
22
|
Manning JA, Shah SS, Nikolic A, Henshall TL, Khew-Goodall Y, Kumar S. The ubiquitin ligase NEDD4-2/NEDD4L regulates both sodium homeostasis and fibrotic signaling to prevent end-stage renal disease. Cell Death Dis 2021; 12:398. [PMID: 33854040 PMCID: PMC8046789 DOI: 10.1038/s41419-021-03688-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Kidney disease progression can be affected by Na+ abundance. A key regulator of Na+ homeostasis is the ubiquitin ligase NEDD4-2 and its deficiency leads to increased Na+ transport activity and salt-sensitive progressive kidney damage. However, the mechanisms responsible for high Na+ induced damage remain poorly understood. Here we show that a high Na+ diet compromised kidney function in Nedd4-2-deficient mice, indicative of progression toward end-stage renal disease. Injury was characterized by enhanced tubule dilation and extracellular matrix accumulation, together with sustained activation of both Wnt/β-catenin and TGF-β signaling. Nedd4-2 knockout in cortical collecting duct cells also activated these pathways and led to epithelial-mesenchymal transition. Furthermore, low dietary Na+ rescued kidney disease in Nedd4-2-deficient mice and silenced Wnt/β-catenin and TGF-β signaling. Our study reveals the important role of NEDD4-2-dependent ubiquitination in Na+ homeostasis and protecting against aberrant Wnt/β-catenin/TGF-β signaling in progressive kidney disease.
Collapse
Affiliation(s)
- Jantina A. Manning
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| | - Sonia S. Shah
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| | - Andrej Nikolic
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| | - Tanya L. Henshall
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| | - Yeesim Khew-Goodall
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| | - Sharad Kumar
- grid.1026.50000 0000 8994 5086Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, 5001 Australia
| |
Collapse
|
23
|
Orr-Burks N, Murray J, Todd KV, Bakre A, Tripp RA. G-Protein-Coupled Receptor and Ion Channel Genes Used by Influenza Virus for Replication. J Virol 2021; 95:e02410-20. [PMID: 33536179 PMCID: PMC8104092 DOI: 10.1128/jvi.02410-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Influenza virus causes epidemics and sporadic pandemics resulting in morbidity, mortality, and economic losses. Influenza viruses require host genes to replicate. RNA interference (RNAi) screens can identify host genes coopted by influenza virus for replication. Targeting these proinfluenza genes can provide therapeutic strategies to reduce virus replication. Nineteen proinfluenza G-protein-coupled receptor (GPCR) and 13 proinfluenza ion channel genes were identified in human lung (A549) cells by use of small interfering RNAs (siRNAs). These proinfluenza genes were authenticated by testing influenza virus A/WSN/33-, A/CA/04/09-, and B/Yamagata/16/1988-infected A549 cells, resulting in the validation of 16 proinfluenza GPCR and 5 proinfluenza ion channel genes. These findings showed that several GPCR and ion channel genes are needed for the production of infectious influenza virus. These data provide potential targets for the development of host-directed therapeutic strategies to impede the influenza virus productive cycle so as to limit infection.IMPORTANCE Influenza epidemics result in morbidity and mortality each year. Vaccines are the most effective preventive measure but require annual reformulation, since a mismatch of vaccine strains can result in vaccine failure. Antiviral measures are desirable particularly when vaccines fail. In this study, we used RNAi screening to identify several GPCR and ion channel genes needed for influenza virus replication. Understanding the host genes usurped by influenza virus during viral replication can help identify host genes that can be targeted for drug repurposing or for the development of antiviral drugs. The targeting of host genes is refractory to drug resistance generated by viral mutations, as well as providing a platform for the development of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Nichole Orr-Burks
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Kyle V Todd
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Abhijeet Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
24
|
Pierandrei S, Truglio G, Ceci F, Del Porto P, Bruno SM, Castellani S, Conese M, Ascenzioni F, Lucarelli M. DNA Methylation Patterns Correlate with the Expression of SCNN1A, SCNN1B, and SCNN1G (Epithelial Sodium Channel, ENaC) Genes. Int J Mol Sci 2021; 22:ijms22073754. [PMID: 33916525 PMCID: PMC8038451 DOI: 10.3390/ijms22073754] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
The interplay between the cystic fibrosis transmembrane conductance regulator (CFTR) and the epithelial sodium channel (ENaC) in respiratory epithelia has a crucial role in the pathogenesis of cystic fibrosis (CF). The comprehension of the mechanisms of transcriptional regulation of ENaC genes is pivotal to better detail the pathogenic mechanism and the genotype-phenotype relationship in CF, as well as to realize therapeutic approaches based on the transcriptional downregulation of ENaC genes. Since we aimed to study the epigenetic transcriptional control of ENaC genes, an assessment of their expression and DNA methylation patterns in different human cell lines, nasal brushing samples, and leucocytes was performed. The mRNA expression of CFTR and ENaC subunits α, β and γ (respectively SCNN1A, SCNN1B, and SCNN1G genes) was studied by real time PCR. DNA methylation of 5'-flanking region of SCNN1A, SCNN1B, and SCNN1G genes was studied by HpaII/PCR. The levels of expression and DNA methylation of ENaC genes in the different cell lines, brushing samples, and leukocytes were very variable. The DNA regions studied of each ENaC gene showed different methylation patterns. A general inverse correlation between expression and DNA methylation was evidenced. Leukocytes showed very low expression of all the 3 ENaC genes corresponding to a DNA methylated pattern. The SCNN1A gene resulted to be the most expressed in some cell lines that, accordingly, showed a completely demethylated pattern. Coherently, a heavy and moderate methylated pattern of, respectively, SCNN1B and SCNN1G genes corresponded to low levels of expression. As exceptions, we found that dexamethasone treatment appeared to stimulate the expression of all the 3 ENaC genes, without an evident modulation of the DNA methylation pattern, and that in nasal brushing a considerable expression of all the 3 ENaC genes were found despite an apparent methylated pattern. At least part of the expression modulation of ENaC genes seems to depend on the DNA methylation patterns of specific DNA regions. This points to epigenetics as a controlling mechanism of ENaC function and as a possible therapeutic approach for CF.
Collapse
Affiliation(s)
- Silvia Pierandrei
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Roma, Italy; (S.P.); (G.T.); (F.C.); (S.M.B.)
| | - Gessica Truglio
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Roma, Italy; (S.P.); (G.T.); (F.C.); (S.M.B.)
| | - Fabrizio Ceci
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Roma, Italy; (S.P.); (G.T.); (F.C.); (S.M.B.)
| | - Paola Del Porto
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, Via dei Sardi 70, 00185 Roma, Italy;
| | - Sabina Maria Bruno
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Roma, Italy; (S.P.); (G.T.); (F.C.); (S.M.B.)
| | - Stefano Castellani
- Department of Biomedical Sciences and Human Oncology, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli 121, 71122 Foggia, Italy;
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, Via dei Sardi 70, 00185 Roma, Italy;
- Correspondence: (F.A.); (M.L.)
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Roma, Italy; (S.P.); (G.T.); (F.C.); (S.M.B.)
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Viale Regina Elena 291, 00161 Roma, Italy
- Correspondence: (F.A.); (M.L.)
| |
Collapse
|
25
|
Bewick GS, Banks RW. Mechanotransduction channels in proprioceptive sensory nerve terminals: still an open question? CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Yadav S, Shaughnessy CA, Zeitlin PL, Bratcher PE. Downregulation of epithelial sodium channel (ENaC) activity in human airway epithelia after low temperature incubation. BMJ Open Respir Res 2021; 8:8/1/e000861. [PMID: 33622672 PMCID: PMC7907861 DOI: 10.1136/bmjresp-2020-000861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/28/2021] [Accepted: 02/13/2021] [Indexed: 11/12/2022] Open
Abstract
Introduction The incubation of airway epithelia cells at low temperatures is a common in vitro experimental approach used in the field of cystic fibrosis (CF) research to thermo-stabilise F508del-CFTR and increase its functional expression. Given that the airway epithelium includes numerous ion transporters other than CFTR, we hypothesised that there was an impact of low temperature incubation on CFTR-independent ionoregulatory mechanisms in airway epithelia derived from individuals with and without CF. Methods After differentiation at the air–liquid interface, nasal epithelia were incubated at either 37°C or 29°C (low temperature) for 48 hours prior to analysis in an Ussing chamber. Results While F508del-CFTR activity was increased after low temperature incubation, activity of CFTR in non-CF epithelia was unchanged. Importantly, cultures incubated at 29°C demonstrated decreased transepithelial potential difference (TEPD) and short-circuit currents (Isc) at baseline. The predominant factor contributing to the reduced baseline TEPD and Isc in 29°C cultures was the reduced activity of the epithelial sodium channel (ENaC), evidenced by a reduced responsiveness to amiloride. This effect was observed in cells derived from both non-CF and CF donors. Discussion Significant transcriptional downregulation of ENaC subunits β and γ were observed, which may partially explain the decreased ENaC activity. We speculate that low temperature incubation may be a useful experimental paradigm to reduce ENaC activity in in vitro epithelial cultures.
Collapse
Affiliation(s)
- Sangya Yadav
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | | | - Pamela L Zeitlin
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Preston E Bratcher
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA .,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
27
|
Cheung TT, Geda AC, Ware AW, Rasulov SR, Tenci P, Hamilton KL, McDonald FJ. Retromer is involved in epithelial Na+ channel trafficking. Am J Physiol Renal Physiol 2020; 319:F895-F907. [DOI: 10.1152/ajprenal.00198.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The epithelial Na+ channel (ENaC) located at the apical membrane in many epithelia is the rate-limiting step for Na+ reabsorption. Tight regulation of the plasma membrane population of ENaC is required, as hypertension or hypotension may result if too many or too few ENaCs are present. Endocytosed ENaC travels to the early endosome and is then either trafficked to the lysosome for degradation or recycled back to the plasma membrane. Recently, the retromer recycling complex, located at the early endosome, has been implicated in plasma membrane protein recycling pathways. We hypothesized that the retromer is required for recycling of ENaC. Stabilization of retromer function with the retromer stabilizing chaperone R55 increased ENaC current, whereas knockdown or overexpression of individual retromer and associated proteins altered ENaC current and cell surface population of ENaC. KIBRA was identified as an ENaC-binding protein allowing ENaC to link to sorting nexin 4 to alter ENaC trafficking. Knockdown of the retromer-associated cargo-binding sorting nexin 27 protein did not alter ENaC current, whereas CCDC22, a CCC-complex protein, coimmunoprecipitated with ENaC, and CCDC22 knockdown decreased ENaC current and population at the cell surface. Together, our results confirm that retromer and the CCC complex play a role in recycling of ENaC to the plasma membrane.
Collapse
Affiliation(s)
- Tanya T. Cheung
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Anna C. Geda
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Adam W. Ware
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sahib R. Rasulov
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Polly Tenci
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kirk L. Hamilton
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Fiona J. McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
28
|
Muhanna D, Arnipalli SR, Kumar SB, Ziouzenkova O. Osmotic Adaptation by Na +-Dependent Transporters and ACE2: Correlation with Hemostatic Crisis in COVID-19. Biomedicines 2020; 8:E460. [PMID: 33142989 PMCID: PMC7693583 DOI: 10.3390/biomedicines8110460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 symptoms, including hypokalemia, hypoalbuminemia, ageusia, neurological dysfunctions, D-dimer production, and multi-organ microthrombosis reach beyond effects attributed to impaired angiotensin-converting enzyme 2 (ACE2) signaling and elevated concentrations of angiotensin II (Ang II). Although both SARS-CoV (Severe Acute Respiratory Syndrome Coronavirus) and SARS-CoV-2 utilize ACE2 for host entry, distinct COVID-19 pathogenesis coincides with the acquisition of a new sequence, which is homologous to the furin cleavage site of the human epithelial Na+ channel (ENaC). This review provides a comprehensive summary of the role of ACE2 in the assembly of Na+-dependent transporters of glucose, imino and neutral amino acids, as well as the functions of ENaC. Data support an osmotic adaptation mechanism in which osmotic and hemostatic instability induced by Ang II-activated ENaC is counterbalanced by an influx of organic osmolytes and Na+ through the ACE2 complex. We propose a paradigm for the two-site attack of SARS-CoV-2 leading to ENaC hyperactivation and inactivation of the ACE2 complex, which collapses cell osmolality and leads to rupture and/or necrotic death of swollen pulmonary, endothelial, and cardiac cells, thrombosis in infected and non-infected tissues, and aberrant sensory and neurological perception in COVID-19 patients. This dual mechanism employed by SARS-CoV-2 calls for combinatorial treatment strategies to address and prevent severe complications of COVID-19.
Collapse
Affiliation(s)
| | | | | | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (D.M.); (S.R.A.); (S.B.K.)
| |
Collapse
|
29
|
Meyer EA, Franklin CE, Cramp RL. Physiological and morphological correlates of extreme acid tolerance in larvae of the acidophilic amphibian Litoria cooloolensis. J Comp Physiol B 2020; 191:159-171. [PMID: 33001282 DOI: 10.1007/s00360-020-01316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 09/03/2020] [Accepted: 09/20/2020] [Indexed: 12/01/2022]
Abstract
The Cooloola sedgefrog (Litoria cooloolensis) is one of a number of frog species endemic to the coastal sandy lowlands of east Australia exhibiting remarkable tolerance to dilute waters of low pH (< pH 3.5). To investigate the physiological and morphological underpinnings of acid tolerance in L. cooloolensis larvae, we compared Na+ balance, uptake and efflux rates, and gill and skin morphology in larvae reared in circum-neutral (pH 6.5) and pH 3.5 water. We hypothesised that L. cooloolensis larvae would be more resistant to ionregulatory disturbance and epithelial damage at low pH relative to acid-sensitive species. Net Na+ flux rates were not significantly different from zero in L. cooloolensis larvae reared at pH 3.5 and in acid-naïve animals maintained in pH 6.5 water. Animals reared at pH 6.5 and acutely exposed to pH 3.5, however, exhibited a net loss of Na+ due to inhibition of Na+ uptake. In contrast, L. cooloolensis larvae reared at pH 3.5 maintained Na+ balance at pH 3.5 and did not exhibit inhibition of Na+ uptake at this pH. Investigation of Na+ transport kinetics and the morphology of the gills and integument suggests tolerance of L. cooloolensis larvae to low pH may be attributed to a high capacity for branchial Na+ uptake, increased tight junction length and elevated mucus production at the gills and integument. These factors confer resistance to acid damage and disruption of ionic homeostasis which would otherwise result in the death of amphibian larvae exposed to waters of pH 4.0 and less.
Collapse
Affiliation(s)
- Edward A Meyer
- School of Biological Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Craig E Franklin
- School of Biological Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Rebecca L Cramp
- School of Biological Sciences, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
30
|
Rico-Mesa JS, White A, Ahmadian-Tehrani A, Anderson AS. Mineralocorticoid Receptor Antagonists: a Comprehensive Review of Finerenone. Curr Cardiol Rep 2020; 22:140. [PMID: 32910349 DOI: 10.1007/s11886-020-01399-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW We aim to review the mechanism of action and safety profile of mineralocorticoid receptor antagonists (MRAs) and discuss the differences between selective and non-selective MRAs. More specifically, finerenone is a new medication that is currently under investigation for its promising cardiovascular and nephrological effects. RECENT FINDINGS MRAs are well known for their utility in treating heart failure, refractory hypertension, and diverse nephropathies, namely, diabetic nephropathy. As their name denotes, MRAs inhibit the action of aldosterone at the mineralocorticoid receptor, preventing receptor activation. This prevents remodeling, decreases inflammation, and improves proteinuria. There are not significant differences in outcomes between selective and non-selective MRAs. A new selective MRA named finerenone (originally BAY 94-8862) has shown promising results in several trials (ARTS-HF and ARTS-DN) and smaller studies. Finerenone may have a dose-dependent benefit over older MRAs, decreasing rates of albuminuria and levels of BNP and NT-ProBNP without causing a significant increase in serum potassium levels. This medication is not yet approved as it is still in phase 3 clinical trials (FIGARO-DKD and FIDELIO-DKD trials). MRAs are beneficial in several disease states. Newer medications, such as finerenone, should be considered in patients with heart failure and diabetic nephropathy who may benefit from a reduction in albuminuria and BNP/NT-ProBNP. Data surrounding finerenone are limited to date. However, results from ongoing clinical trials, as well as new trials to evaluate use in other pathologies, could validate the implementation of this medication in daily practice.
Collapse
Affiliation(s)
- Juan Simon Rico-Mesa
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Averi White
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Ashkan Ahmadian-Tehrani
- Department of Medicine, Division of Internal Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Allen S Anderson
- Department of Medicine, Division of Cardiovascular Diseases, University of Texas Health San Antonio, 7703 Floyd Curl Drive, MC 7872, San Antonio, TX, 78229, USA.
| |
Collapse
|
31
|
The physiological and molecular mechanisms to maintain water and salt homeostasis in response to high salt intake in Mongolian gerbils (Meriones unguiculatus). J Comp Physiol B 2020; 190:641-654. [PMID: 32556536 DOI: 10.1007/s00360-020-01287-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022]
Abstract
Desert rodents are faced with many challenges such as high dietary salt in their natural habitats and they have evolved abilities to conserve water and tolerate salt. However, the physiological and molecular mechanisms involved in water and salt balances in desert rodents are unknown. We hypothesized that desert rodents regulated water and salt balances by altering the expression of AQP2 and α-ENaC in the kidney. Mongolian gerbils (Meriones unguiculatus), a desert species, were acclimated to drinking water with different salt contents: (0, control; 4% NaCl, moderate salt, MS; 8% NaCl, high salt, HS) for 4 weeks. The gerbils drinking salty water had lower body mass, food intake, water intake, metabolic water production and urine volume. The HS gerbils increased the expression of arginine vasopressin (AVP) in the hypothalamus, and also enhanced the expression of AQP2 and cAMP/PKA/CREB signaling pathway in the kidney. In addition, these gerbils reduced serum aldosterone levels and α-ENaC expression in the kidney. Creatinine clearance was lower in the HS group than that in the control group, but serum and urine creatinine levels did not change. These data indicate that desert rodents rely on AVP-dependent upregulation of AQP2 and aldosterone-dependent downregulation of α-ENaC in the kidney to promote water reabsorption and sodium excretion under high salt intake.
Collapse
|
32
|
Anand P, Puranik A, Aravamudan M, Venkatakrishnan AJ, Soundararajan V. SARS-CoV-2 strategically mimics proteolytic activation of human ENaC. eLife 2020; 9:58603. [PMID: 32452762 PMCID: PMC7343387 DOI: 10.7554/elife.58603] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Molecular mimicry is an evolutionary strategy adopted by viruses to exploit the host cellular machinery. We report that SARS-CoV-2 has evolved a unique S1/S2 cleavage site, absent in any previous coronavirus sequenced, resulting in the striking mimicry of an identical FURIN-cleavable peptide on the human epithelial sodium channel α-subunit (ENaC-α). Genetic alteration of ENaC-α causes aldosterone dysregulation in patients, highlighting that the FURIN site is critical for activation of ENaC. Single cell RNA-seq from 66 studies shows significant overlap between expression of ENaC-α and the viral receptor ACE2 in cell types linked to the cardiovascular-renal-pulmonary pathophysiology of COVID-19. Triangulating this cellular characterization with cleavage signatures of 178 proteases highlights proteolytic degeneracy wired into the SARS-CoV-2 lifecycle. Evolution of SARS-CoV-2 into a global pandemic may be driven in part by its targeted mimicry of ENaC-α, a protein critical for the homeostasis of airway surface liquid, whose misregulation is associated with respiratory conditions. Viruses hijack the cellular machinery of humans to infect their cells and multiply. The virus causing the global COVID-19 pandemic, SARS-CoV-2, is no exception. Identifying which proteins in human cells the virus co-opts is crucial for developing new ways to diagnose, prevent and treat COVID-19 infections. SARS-CoV-2 is covered in spike-shaped proteins, which the virus uses to gain entry into cells. First, the spikes bind to a protein called ACE2, which is found on the cells that line the respiratory tract and lungs. SARS-CoV-2 then exploits enzymes called proteases to cut, or cleave, its spikes at a specific site which allows the virus to infiltrate the host cell. Proteases identify which proteins to target based on the sequence of amino acids – the building blocks of proteins – at the cleavage site. However, it remained unclear which human proteases SARS-CoV-2 co-opts and whether its cut site is similar to human proteins. Now, Anand et al. show that the spike proteins on SARS-CoV-2 may have the same sequence of amino acids at its cut site as a human epithelial channel protein called ENaC-α. This channel is important for maintaining the balance of salt and water in many organs including the lungs. Further analyses showed that ENaC-α is often found in the same types of human lung and respiratory tract cells as ACE2. This suggests that SARS-CoV-2 may use the same proteases that cut ENaC-α to get inside human respiratory cells. It is possible that by hijacking the cutting mechanism for ENaC-α, SARS-CoV-2 interferes with the balance of salt and water in the lungs of COVID-19 patients. This may help explain why the virus causes severe respiratory symptoms. However, more studies are needed to confirm that the proteases that cut ENaC-α also cut the spike proteins on SARS-CoV-2, and how this affects the respiratory health of COVID-19 patients.
Collapse
|
33
|
Effects of syntaxins 2, 3, and 4 on rat and human epithelial sodium channel (ENaC) in Xenopus laevis oocytes. Pflugers Arch 2020; 472:461-471. [PMID: 32221667 PMCID: PMC7165155 DOI: 10.1007/s00424-020-02365-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 01/20/2023]
Abstract
Syntaxins are SNARE proteins and may play a role in epithelial sodium channel (ENaC) trafficking. The aim of the present study was to investigate the effects of syntaxin 2 (STX2), syntaxin 3 (STX3), and syntaxin 4 (STX4) on rat (rENaC) and human ENaC (hENaC). Co-expression of rENaC and STX3 or STX4 in Xenopus laevis oocytes increased amiloride-sensitive whole-cell currents (ΔIami) on average by 50% and 135%, respectively, compared to oocytes expressing rENaC alone. In contrast, STX2 had no significant effect on rENaC. Similar to its effect on rENaC, STX3 stimulated hENaC by 48%. In contrast, STX2 and STX4 inhibited hENaC by 51% and 44%, respectively. Using rENaC carrying a FLAG tag in the extracellular loop of the β-subunit, we demonstrated that the stimulatory effects of STX3 and STX4 on ΔIami were associated with an increased expression of the channel at the cell surface. Co-expression of STX3 or STX4 did not significantly alter the degree of proteolytic channel activation by chymotrypsin. STX3 had no effect on the inhibition of rENaC by brefeldin A, and the stimulatory effect of STX3 was preserved in the presence of dominant negative Rab11. This indicates that the stimulatory effect of STX3 is not mediated by inhibiting channel retrieval or by stimulating fusion of recycling endosomes. Our results suggest that the effects of syntaxins on ENaC are isoform and species dependent. Furthermore, our results demonstrate that STX3 increases ENaC expression at the cell surface, probably by enhancing insertion of vesicles carrying newly synthesized channels.
Collapse
|
34
|
Enns CB, Keith BA, Challa N, Harding JCS, Loewen ME. Impairment of electroneutral Na + transport and associated downregulation of NHE3 contributes to the development of diarrhea following in vivo challenge with Brachyspira spp. Am J Physiol Gastrointest Liver Physiol 2020; 318:G288-G297. [PMID: 31760765 PMCID: PMC7052572 DOI: 10.1152/ajpgi.00011.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The effect of Brachyspira hyodysenteriae and Brachyspira hampsonii spirochetosis on Na+ transport was assessed in the colon to determine its contribution to diarrheal disease in pigs following experimental infection. Electrogenic and electroneutral Na+ absorption was assessed in Ussing chambers by radiolabeled 22Na flux and pharmacological inhibitory studies. Basal radiolabeled 22Na flux experiments revealed that mucosal-to-serosal flux (Jms) was significantly impaired in B. hyodysenteriae and B. hampsonii-diseased pigs. Inhibition of epithelial sodium channel via amiloride did not significantly reduce electrogenic short-circuit current (Isc) in the proximal, apex, and distal colonic segments of diseased pigs over control pigs, suggesting that a loss of electroneutral Na+ absorption is responsible for diarrheal development. These findings were further supported by significant downregulation of Na+/H+ exchanger (NHE1, NHE2, and NHE3) mRNA expression in the proximal, apex, and distal colonic segments paired with decreased protein expression of the critical NHE3 isoform. The decrease in NHE3 mRNA expression appears not to be attributed to the host's cytokine response as human IL-1α did not modify NHE3 mRNA expression in Caco-2 cells. However, a whole cell B. hampsonii lysate significantly downregulated NHE3 mRNA expression and significantly increased p38 phosphorylation in Caco-2 cells. Together these findings provide a likely mechanism for the spirochete-induced malabsorptive diarrhea, indicated by a decrease in electroneutral Na+ absorption in the porcine colon due to Brachyspira's ability to inhibit NHE3 transcription, resulting in diarrheal disease.NEW & NOTEWORTHY This research demonstrates that diarrheal disease caused by two infectious spirochete spp. is a result of impaired electroneutral Na+ absorption via Na+/H+ exchanger 3 (NHE3) in the porcine colon. Our findings suggest that the decrease in NHE3 mRNA and protein is not likely a result of the host's cytokine response. Rather, it appears that these two Brachyspira spp. directly inhibit the transcription and translation of NHE3, resulting in the development of diarrhea.
Collapse
Affiliation(s)
- Cole B. Enns
- 1Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Brandon A. Keith
- 1Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Nitin Challa
- 1Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John C. S. Harding
- 2Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Matthew E. Loewen
- 1Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
35
|
Bukhari AAS, Zhang X, Li M, Zhao A, Dong H, Liang X. Cofilin participates in regulating alpha-epithelial sodium channel by interaction with 14-3-3 isoforms. J Biomed Res 2020; 34:351-360. [PMID: 32981895 PMCID: PMC7540242 DOI: 10.7555/jbr.34.20190155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Renal epithelial sodium channel (ENaC) plays a crucial role in maintaining homeostasis and sodium absorption. While insulin participates in controlling sodium transport across the renal epithelium, the underlying molecular mechanism remain unclear. In this study, we found that insulin increased the expression and function of alpha-epithelial sodium channel (α-ENaC) as well as phosphorylation of cofilin, a family of actin-binding proteins which disassembles actin filaments, in mouse cortical collecting duct (mpkCCDc14) cells. The wild-type (WT) cofilin and its constitutively phosphorylated form (S3D), but not its constitutively non-phosphorylable form (S3A), contributed to the elevated expression on α-ENaC. Overexpression of 14-3-3ε, β, or γ increased the expression of α-ENaC and cofilin phosphorylation, which was blunted by knockdown of 14-3-3ε, β, or γ. Moreover, it was found that insulin increased the interaction between cofilin and 14-3-3 isoforms, which indicated relevance of 14-3-3 isoforms with cofilin. Furthermore, LIMK1/SSH1 pathway was involved in regulation of cofilin and α-ENaC expression by insulin. The results from this work indicate that cofilin participates in the regulation of α-ENaC by interaction with 14-3-3 isoforms.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiubin Liang
- Department of Pathophysiology;Department of Nephrology, the Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
36
|
Kleyman TR, Eaton DC. Regulating ENaC's gate. Am J Physiol Cell Physiol 2020; 318:C150-C162. [PMID: 31721612 PMCID: PMC6985836 DOI: 10.1152/ajpcell.00418.2019] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023]
Abstract
Epithelial Na+ channels (ENaCs) are members of a family of cation channels that function as sensors of the extracellular environment. ENaCs are activated by specific proteases in the biosynthetic pathway and at the cell surface and remove embedded inhibitory tracts, which allows channels to transition to higher open-probability states. Resolved structures of ENaC and an acid-sensing ion channel revealed highly organized extracellular regions. Within the periphery of ENaC subunits are unique domains formed by antiparallel β-strands containing the inhibitory tracts and protease cleavage sites. ENaCs are inhibited by Na+ binding to specific extracellular site(s), which promotes channel transition to a lower open-probability state. Specific inositol phospholipids and channel modification by Cys-palmitoylation enhance channel open probability. How these regulatory factors interact in a concerted manner to influence channel open probability is an important question that has not been resolved. These various factors are reviewed, and the impact of specific factors on human disorders is discussed.
Collapse
Affiliation(s)
- Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, and Departments of Cell Biology and of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas C Eaton
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
37
|
Dietary sodium modulates nephropathy in Nedd4-2-deficient mice. Cell Death Differ 2019; 27:1832-1843. [PMID: 31802037 PMCID: PMC7244563 DOI: 10.1038/s41418-019-0468-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/05/2022] Open
Abstract
Salt homeostasis is maintained by tight control of Na+ filtration and reabsorption. In the distal part of the nephron the ubiquitin protein ligase Nedd4-2 regulates membrane abundance and thus activity of the epithelial Na+ channel (ENaC), which is rate-limiting for Na+ reabsorption. Nedd4-2 deficiency in mouse results in elevated ENaC and nephropathy, however the contribution of dietary salt to this has not been characterized. In this study we show that high dietary Na+ exacerbated kidney injury in Nedd4-2-deficient mice, significantly perturbing normal postnatal nephrogenesis and resulting in multifocal areas of renal dysplasia, increased markers of kidney injury and a decline in renal function. In control mice, high dietary Na+ resulted in reduced levels of ENaC. However, Nedd4-2-deficient kidneys maintained elevated ENaC even after high dietary Na+, suggesting that the inability to efficiently downregulate ENaC is responsible for the salt-sensitivity of disease. Importantly, low dietary Na+ significantly ameliorated nephropathy in Nedd4-2-deficient mice. Our results demonstrate that due to dysregulation of ENaC, kidney injury in Nedd4-2-deficient mice is sensitive to dietary Na+, which may have implications in the management of disease in patients with kidney disease.
Collapse
|
38
|
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
|
39
|
Ware AW, Rasulov SR, Cheung TT, Lott JS, McDonald FJ. Membrane trafficking pathways regulating the epithelial Na + channel. Am J Physiol Renal Physiol 2019; 318:F1-F13. [PMID: 31657249 DOI: 10.1152/ajprenal.00277.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Renal Na+ reabsorption, facilitated by the epithelial Na+ channel (ENaC), is subject to multiple forms of control to ensure optimal body blood volume and pressure through altering both the ENaC population and activity at the cell surface. Here, the focus is on regulating the number of ENaCs present in the apical membrane domain through pathways of ENaC synthesis and targeting to the apical membrane as well as ENaC removal, recycling, and degradation. Finally, the mechanisms by which ENaC trafficking pathways are regulated are summarized.
Collapse
Affiliation(s)
- Adam W Ware
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sahib R Rasulov
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tanya T Cheung
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - J Shaun Lott
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Fiona J McDonald
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
40
|
Casini S, Albesa M, Wang Z, Portero V, Ross-Kaschitza D, Rougier JS, Marchal GA, Chung WK, Bezzina CR, Abriel H, Remme CA. Functional Consequences of the SCN5A-p.Y1977N Mutation within the PY Ubiquitylation Motif: Discrepancy between HEK293 Cells and Transgenic Mice. Int J Mol Sci 2019; 20:ijms20205033. [PMID: 31614475 PMCID: PMC6829230 DOI: 10.3390/ijms20205033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/04/2019] [Accepted: 10/06/2019] [Indexed: 01/25/2023] Open
Abstract
Dysfunction of the cardiac sodium channel Nav1.5 (encoded by the SCN5A gene) is associated with arrhythmias and sudden cardiac death. SCN5A mutations associated with long QT syndrome type 3 (LQT3) lead to enhanced late sodium current and consequent action potential (AP) prolongation. Internalization and degradation of Nav1.5 is regulated by ubiquitylation, a post-translational mechanism that involves binding of the ubiquitin ligase Nedd4-2 to a proline-proline-serine-tyrosine sequence of Nav1.5, designated the PY-motif. We investigated the biophysical properties of the LQT3-associated SCN5A-p.Y1977N mutation located in the Nav1.5 PY-motif, both in HEK293 cells as well as in newly generated mice harboring the mouse homolog mutation Scn5a-p.Y1981N. We found that in HEK293 cells, the SCN5A-p.Y1977N mutation abolished the interaction between Nav1.5 and Nedd4-2, suppressed PY-motif-dependent ubiquitylation of Nav1.5, and consequently abrogated Nedd4-2 induced sodium current (INa) decrease. Nevertheless, homozygous mice harboring the Scn5a-p.Y1981N mutation showed no electrophysiological alterations nor changes in AP or (late) INa properties, questioning the in vivo relevance of the PY-motif. Our findings suggest the presence of compensatory mechanisms, with additional, as yet unknown, factors likely required to reduce the “ubiquitylation reserve” of Nav1.5. Future identification of such modulatory factors may identify potential triggers for arrhythmias and sudden cardiac death in the setting of LQT3 mutations.
Collapse
Affiliation(s)
- Simona Casini
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 Amsterdam, The Netherlands.
| | - Maxime Albesa
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Zizun Wang
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Vincent Portero
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 Amsterdam, The Netherlands.
| | - Daniela Ross-Kaschitza
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Jean-Sébastien Rougier
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Gerard A Marchal
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 Amsterdam, The Netherlands.
| | - Wendy K Chung
- Departments of Pediatrics & Medicine, Columbia University Medical Center, 1150 St Nicholas Avenue, New York, NY 10032, USA.
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 Amsterdam, The Netherlands.
| | - Hugues Abriel
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Gumbel JH, Montgomery LR, Yang CB, Hubscher CH. Activity-Based Training Reverses Spinal Cord Injury-Induced Changes in Kidney Receptor Densities and Membrane Proteins. J Neurotrauma 2019; 37:555-563. [PMID: 31456470 DOI: 10.1089/neu.2019.6670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Complications in upper and lower urinary function arise after spinal cord injury (SCI), which creates a significant impact on quality of life for those affected. One upper urinary complication is SCI-induced polyuria, or the overproduction of urine, of which the underlying mechanisms have yet to be elucidated. Activity-based training (ABT) has been utilized in both animal and clinical settings as a rehabilitative therapy to improve many issues that arise after SCI, including more recently urogenital function. The goal of the current study was to identify potential mechanisms contributing to previously identified improvements in polyuria with ABT, using a male rat moderate-severe spinal contusion model. Although ABT had no significant effect on reversing injury-induced alterations of serum arginine vasopressin and urinary atrial natriuretic peptide levels, there was a dramatic effect upon the receptors of these fluid balance hormones (vasopressin receptor 2 and natriuretic peptide A receptor), as well as kidney aquaporin 2 and sodium channels. ABT changes in densities of key receptors and kidney membrane proteins involved in fluid balance after chronic SCI support the likelihood of multiple mechanisms through which exercise can positively influence urinary tract function after SCI. By understanding the mechanisms, amount, and timing regarding how ABT improves different aspects of urinary function, more targeted training strategies can be developed to optimize the functional gains within the SCI population.
Collapse
Affiliation(s)
- Jason H Gumbel
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky
| | - Lynnette R Montgomery
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky.,Kentucky Spinal Cord Injury Rehabilitation Center, University of Louisville, Louisville, Kentucky
| | - Cui Bo Yang
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky.,Kentucky Spinal Cord Injury Rehabilitation Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
42
|
Mezger A, Ruland A, Ruef P. Pseudohypoaldosteronismus Typ I. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-019-00787-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
Klemens CA, Brands MW, Staruschenko A. Postprandial effects on electrolyte homeostasis in the kidney. Am J Physiol Renal Physiol 2019; 317:F1405-F1408. [PMID: 31566434 DOI: 10.1152/ajprenal.00350.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Insulin is known to be an important regulator of a number of different channels and transporters in the kidney, but its role in the kidney to prevent Na+ and volume loss during the osmotic load after a meal has only recently been validated. With increasing numbers of people suffering from diabetes and hypertension, furthering our understanding of insulin signaling and renal Na+ handling in both normal and diseased states is essential for improving patient treatments and outcomes. The present review is focused on postprandial effects on Na+ reabsorption in the kidney and the role of the epithelial Na+ channels as an important channel contributing to insulin-mediated Na+ reclamation.
Collapse
Affiliation(s)
- Christine A Klemens
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
44
|
Olivença DV, Fonseca LL, Voit EO, Pinto FR. Thickness of the airway surface liquid layer in the lung is affected in cystic fibrosis by compromised synergistic regulation of the ENaC ion channel. J R Soc Interface 2019; 16:20190187. [PMID: 31455163 DOI: 10.1098/rsif.2019.0187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The lung epithelium is lined with a layer of airway surface liquid (ASL) that is crucial for healthy lung function. ASL thickness is controlled by two ion channels: epithelium sodium channel (ENaC) and cystic fibrosis (CF) transmembrane conductance regulator (CFTR). Here, we present a minimal mathematical model of ENaC, CFTR and ASL regulation that sheds light on the control of ENaC by the short palate lung and nasal epithelial clone 1 (SPLUNC1) protein and by phosphatidylinositol 4,5-biphosphate (PI(4,5)P2). The model, despite its simplicity, yields a good fit to experimental observations and is an effective tool for exploring the interplay between ENaC, CFTR and ASL. Steady-state data and dynamic information constrain the model's parameters without ambiguities. Testing the hypothesis that PI(4,5)P2 protects ENaC from ubiquitination suggests that this protection does not improve the model results and that the control of the ENaC opening probability by PI(4,5)P2 is sufficient to explain all available data. The model analysis further demonstrates that ASL at the steady state is sensitive to small changes in PI(4,5)P2 abundance, particularly in CF conditions, which suggests that manipulation of phosphoinositide metabolism may promote therapeutic benefits for CF patients.
Collapse
Affiliation(s)
- Daniel V Olivença
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| | - Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Francisco R Pinto
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| |
Collapse
|
45
|
Zhang X, Ge Y, Bukhari AAS, Zhu Q, Shen Y, Li M, Sun H, Su D, Liang X. Estrogen negatively regulates the renal epithelial sodium channel (ENaC) by promoting Derlin-1 expression and AMPK activation. Exp Mol Med 2019; 51:1-12. [PMID: 31113930 PMCID: PMC6529463 DOI: 10.1038/s12276-019-0253-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022] Open
Abstract
The main functions of the epithelial sodium channel (ENaC) in the kidney distal nephron are mediation of sodium and water balance and stabilization of blood pressure. Estrogen has important effects on sodium and water balance and on premenopausal blood pressure, but its role in the regulation of ENaC function is not fully understood. Female Sprague–Dawley rats were treated with 17β-estradiol for 6 weeks following bilateral ovariectomy. Plasma estrogen, aldosterone, creatinine, and electrolytes were analyzed, and α-ENaC and derlin-1 protein expression in the kidney was determined by immunohistochemistry and western blotting. The expression levels of α-ENaC, derlin-1, AMPK, and related molecules were also examined by western blotting and real-time PCR in cultured mouse renal collecting duct (mpkCCDc14) epithelial cells following estrogen treatment. Immunofluorescence and coimmunoprecipitation were performed to detect α-ENaC binding with derlin-1 and α-ENaC ubiquitination. The results demonstrated that the loss of estrogen elevated systolic blood pressure in ovariectomized (OVX) rats. OVX rat kidneys showed increased α-ENaC expression but decreased derlin-1 expression. In contrast, estrogen treatment decreased α-ENaC expression but increased derlin-1 expression in mpkCCDc14 cells. Moreover, estrogen induced α-ENaC ubiquitination by promoting the interaction of α-ENaC with derlin-1 and evoked phosphorylation of AMPK in mpkCCDc14 cells. Our study indicates that estrogen reduces ENaC expression and blood pressure in OVX rats through derlin-1 upregulation and AMPK activation. Estrogen treatment could prove valuable in tackling high blood pressure (hypertension) in postmenopausal women. Long-term healthy blood pressure is linked to the correct regulation of sodium and water levels in the kidneys. The renal epithelial sodium channel (ENaC) is a cellular membrane channel responsible for mediating sodium reabsorption and fluid balance. Liang and co-workers at Nanjing Medical University in Nanjing, China, conducted experiments on postmenopausal rat models, and found that loss of estrogen elevates systolic blood pressure (the pressure during heart muscle contraction), and that the rats had high levels of ENaC expression. Further investigations showed that estrogen treatment restored blood pressure to normal levels by promoting two key proteins involved in cellular membrane health and energy metabolism. This in turn reinstated normal levels of ENaC breakdown in the kidneys, limiting hypertension.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yamei Ge
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | - Qian Zhu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yachen Shen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Min Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hui Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Dongming Su
- Center of Pathology and Clinical Laboratory, Sir Run Run Hospital, Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
46
|
Niu F, Xu X, Zhang R, Sun L, Gan N, Wang A. Ursodeoxycholic acid stimulates alveolar fluid clearance in LPS-induced pulmonary edema via ALX/cAMP/PI3K pathway. J Cell Physiol 2019; 234:20057-20065. [PMID: 30972764 DOI: 10.1002/jcp.28602] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
This study aims to examine the impact of ursodeoxycholic acid (UDCA) on pulmonary edema and explore the underlying molecular mechanisms. The effects of UDCA on pulmonary edema were assessed through hematoxylin and eosin (H&E) staining, lung dry/wet (W/D) ratio, TNF-α/IL-1β levels of bronchoalveolar lavage fluid (BALF), protein expression of epithelial sodium channel (ENaC), and Na+ /K+ -ATPase. Besides, the detailed mechanisms were explored in primary rat alveolar type (AT) II epithelial cells by determining the effects of BOC-2 (ALX [lipoxin A4 receptor] inhibitor), Rp-cAMP (cAMP inhibitor), LY294002 (PI3K inhibitor), and H89 (PKA inhibitor) on the therapeutic effects of UDCA against lipopolysaccharide (LPS)-induced changes. Histological examination suggested that LPS-induced lung injury was obviously attenuated by UDCA. BALF TNF-α/IL-1β levels and lung W/D ratios were decreased by UDCA in LPS model rats. UDCA stimulated alveolar fluid clearance (AFC) though the upregulation of ENaC and Na+ /K+ -ATPase. BOC-2, Rp-cAMP, and LY294002 largely suppressed the therapeutic effects of UDCA. Significant attenuation of pulmonary edema and lung inflammation was revealed in LPS-challenged rats after the UDCA treatment. The therapeutic efficacy of UDCA against LPS was mainly achieved through the ALX/cAMP/PI3K pathway. Our results suggested that UDCA might be a potential drug for the treatment of pulmonary edema induced by LPS.
Collapse
Affiliation(s)
- Fangfang Niu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Rong Zhang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lingling Sun
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ning Gan
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
47
|
Rogers RT, Sun MA, Yue Q, Bao HF, Sands JM, Blount MA, Eaton DC. Lack of urea transporters, UT-A1 and UT-A3, increases nitric oxide accumulation to dampen medullary sodium reabsorption through ENaC. Am J Physiol Renal Physiol 2019; 316:F539-F549. [PMID: 30539654 PMCID: PMC6459308 DOI: 10.1152/ajprenal.00166.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 12/31/2022] Open
Abstract
Although the role of urea in urine concentration is known, the effect of urea handling by the urea transporters (UTs), UT-A1 and UT-A3, on sodium balance remains elusive. Serum and urinary sodium concentration is similar between wild-type mice (WT) and UT-A3 null (UT-A3 KO) mice; however, mice lacking both UT-A1 and UT-A3 (UT-A1/A3 KO) have significantly lower serum sodium and higher urinary sodium. Protein expression of renal sodium transporters is unchanged among all three genotypes. WT, UT-A3 KO, and UT-A1/A3 KO acutely respond to hydrochlorothiazide and furosemide; however, UT-A1/A3 KO fail to show a diuretic or natriuretic response following amiloride administration, indicating that baseline epithelial Na+ channel (ENaC) activity is impaired. UT-A1/A3 KO have more ENaC at the apical membrane than WT mice, and single-channel analysis of ENaC in split-open inner medullary collecting duct (IMCD) isolated in saline shows that ENaC channel density and open probability is higher in UT-A1/A3 KO than WT. UT-A1/A3 KO excrete more urinary nitric oxide (NO), a paracrine inhibitor of ENaC, and inner medullary nitric oxide synthase 1 mRNA expression is ~40-fold higher than WT. Because endogenous NO is unstable, ENaC activity was reassessed in split-open IMCD with the NO donor PAPA NONOate [1-propanamine-3-(2-hydroxy-2-nitroso-1-propylhydrazine)], and ENaC activity was almost abolished in UT-A1/A3 KO. In summary, loss of both UT-A1 and UT-A3 (but not UT-A3 alone) causes elevated medullary NO production and salt wasting. NO inhibition of ENaC, despite elevated apical accumulation of ENaC in UT-A1/A3 KO IMCD, appears to be the main contributor to natriuresis in UT-A1/A3 KO mice.
Collapse
Affiliation(s)
- Richard T Rogers
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Michael A Sun
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Qiang Yue
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Hui-Fang Bao
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Mitsi A Blount
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
48
|
Cheung TT, Ismail NAS, Moir R, Arora N, McDonald FJ, Condliffe SB. Annexin II Light Chain p11 Interacts With ENaC to Increase Functional Activity at the Membrane. Front Physiol 2019; 10:7. [PMID: 30800070 PMCID: PMC6375906 DOI: 10.3389/fphys.2019.00007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 01/07/2019] [Indexed: 11/13/2022] Open
Abstract
The epithelial Na+ channel (ENaC) provides for Na+ absorption in various types of epithelia including the kidney, lung, and colon where ENaC is localized to the apical membrane to enable Na+ entry into the cell. The degree of Na+ entry via ENaC largely depends on the number of active channels localized to the cell membrane, and is tightly controlled by interactions with ubiquitin ligases, kinases, and G-proteins. While regulation of ENaC endocytosis has been well-studied, relatively little is understood of the proteins that govern ENaC exocytosis. We hypothesized that the annexin II light chain, p11, could participate in the transport of ENaC along the exocytic pathway. Our results demonstrate that all three ENaC channel subunits interacted with p11 in an in vitro binding assay. Furthermore, p11 was able to immunoprecipitate ENaC in epithelial cells. Quantitative mass spectrometry of affinity-purified ENaC-p11 complexes recovered several other trafficking proteins including HSP-90 and annexin A6. We also report that p11 exhibits a robust protein expression in cortical collecting duct epithelial cells. However, the expression of p11 in these cells was not influenced by either short-term or long-term exposure to aldosterone. To determine whether the p11 interaction affected ENaC function, we measured amiloride sensitive Na+ currents in Xenopus oocytes or mammalian epithelia co-expressing ENaC and p11 or a siRNA to p11. Results from these experiments showed that p11 significantly augmented ENaC current, whereas knockdown of p11 decreased current. Further, knockdown of p11 reduced ENaC cell surface population suggesting p11 promotes membrane insertion of ENaC. Overall, our findings reveal a novel protein interaction that controls the number of ENaC channels inserted at the membrane via the exocytic pathway.
Collapse
Affiliation(s)
- Tanya T Cheung
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Noor A S Ismail
- Department of Physiology, University of Otago, Dunedin, New Zealand.,Biochemistry Department, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Rachel Moir
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Nikhil Arora
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Fiona J McDonald
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
49
|
Khadijah Ramli NS, Giribabu N, Salleh N. Testosterone enhances expression and functional activity of epithelial sodium channel (ENaC), cystic fibrosis transmembrane regulator (CFTR) and sodium hydrogen exchanger (NHE) in vas deferens of sex-steroid deficient male rats. Steroids 2018; 138:117-133. [PMID: 30003911 DOI: 10.1016/j.steroids.2018.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/19/2022]
Abstract
UNLABELLED Effects of testosterone on expression and functional activity of ENaC, CFTR and NHE in vas deferens were investigated. METHODS Orchidectomized, adult male rats were given 125 and 250 μg/kg/day testosterone subcutaneously, with or without flutamide and finasteride for seven consecutive days. At the end of the treatment, rats were anesthetized and vas deferens were perfused. Changes in vas deferens fluid secretion rate, pH, HCO3-, Cl- and Na+ concentrations were recorded in the presence of amiloride and Cftr inh-172. Rats were then sacrificed and vas deferens were harvested and subjected for molecular biological analysis. RESULTS Testosterone treatment caused the fluid pH and HCO3- concentrations to decrease but secretion rate, Cl- and Na+ concentrations to increase, where upon amiloride administration, the pH and HCO3- concentration increased but Cl- and Na+ concentrations further increased. In testosterone-treated rats, administration of Cftr inh-172 caused all fluid parameters to decrease. In testosterone-treated rats co-administered with flutamide or finasteride, pH and HCO3- concentration increased but fluid secretion rate, Cl- and Na+ concentrations decreased and these parameters were not affected by amiloride or Cftr inh-172 administration. Under testosterone influence, CFTR and γ-ENaC were highly expressed at the apical membrane while NHE-1 and 4 were highly expressed at the basolateral membrane of vas deferens epithelium. Meanwhile, NHE-2 and 3 were highly expressed at the apical membrane. CONCLUSIONS Differential expression of ENaC, CFTR and NHE in vas deferens under testosterone influence indicated the important role of these transporters in creating optimal fluid microenvironment that is essential for preserving male fertility.
Collapse
Affiliation(s)
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
50
|
Dong OM. Excessive dietary sodium intake and elevated blood pressure: a review of current prevention and management strategies and the emerging role of pharmaconutrigenetics. BMJ Nutr Prev Health 2018; 1:7-16. [PMID: 33235949 PMCID: PMC7678480 DOI: 10.1136/bmjnph-2018-000004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Olivia M Dong
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|