1
|
Houser A, Baconguis I. Structural insights into subunit-dependent functional regulation in epithelial sodium channels. Structure 2025; 33:349-362.e4. [PMID: 39667931 PMCID: PMC11805665 DOI: 10.1016/j.str.2024.11.013] [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: 07/01/2024] [Revised: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Epithelial sodium channels (ENaCs) play a crucial role in Na+ reabsorption in mammals. To date, four subunits have been identified-α, β, γ, and δ-believed to form different heteromeric complexes. Currently, only the structure of the αβγ complex is known. To investigate the formation of channels with different subunit compositions and to determine how each subunit contributes to distinct channel properties, we co-expressed human δ, β, and γ. Using single-particle cryoelectron microscopy, we observed three distinct ENaC complexes. The structures unveil a pattern in which β and γ positions are conserved among the different complexes while the α position in αβγ trimer is occupied by either δ or another β. The δ subunit induces structural rearrangements in the γ subunit, which may contribute to the differences in channel activity between αβγ and δβγ channels. These structural changes provide molecular insights into how ENaC subunit composition modulates channel function.
Collapse
Affiliation(s)
- Alexandra Houser
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Isabelle Baconguis
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
2
|
Paudel P, McDonald F, Fronius M. Isolation of Endothelial Cells from Human Internal Mammary Artery. Methods Mol Biol 2025; 2894:21-34. [PMID: 39699807 DOI: 10.1007/978-1-0716-4342-6_3] [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] [Indexed: 12/20/2024]
Abstract
Endothelial cells are a major contributor to cardiovascular diseases. Studying their function and how they influence pathological processes remains an ongoing area of research. Although primary endothelial cells might be readily available from animals, translating results from these studies can be challenging due to species-dependent differences. A common source of human endothelial cells is human umbilical vein endothelial cells, but these cells might not represent the variety of endothelial cells from various vascular beds. Here we describe a protocol for the isolation and cultivation of endothelial cells from human internal mammary artery remains. These remains are commonly available from coronary artery bypass graft surgeries. The described tissue explant method combined with a magnetic sorting process provided a relatively high yield of endothelial cells that were subsequently passaged and used for studies involving mRNA and protein expression analyses, as well as fluorescence-based immunohistochemistry and patch-clamp electrophysiology. This protocol may help to extend the repertoire of studying human endothelial cells to understand their role and function in health and disease.
Collapse
Affiliation(s)
- Puja Paudel
- Department of Physiology, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Fiona McDonald
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin, New Zealand.
- HeartOtago, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
3
|
Wang XP, Srinivasan P, El Hamdaoui M, Blobner BM, Grytz R, Kashlan OB. Varying Selection Pressure for a Na+ Sensing Site in Epithelial Na+ Channel Subunits Reflect Divergent Roles in Na+ Homeostasis. Mol Biol Evol 2024; 41:msae162. [PMID: 39101592 PMCID: PMC11331422 DOI: 10.1093/molbev/msae162] [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: 02/21/2024] [Revised: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024] Open
Abstract
The epithelial Na+ channel (ENaC) emerged early in vertebrates and has played a role in Na+ and fluid homeostasis throughout vertebrate evolution. We previously showed that proteolytic activation of the channel evolved at the water-to-land transition of vertebrates. Sensitivity to extracellular Na+, known as Na+ self-inhibition, reduces ENaC function when Na+ concentrations are high and is a distinctive feature of the channel. A fourth ENaC subunit, δ, emerged in jawed fishes from an α subunit gene duplication. Here, we analyzed 849 α and δ subunit sequences and found that a key Asp in a postulated Na+ binding site was nearly always present in the α subunit, but frequently lost in the δ subunit (e.g. human). Analysis of site evolution and codon substitution rates provide evidence that the ancestral α subunit had the site and that purifying selection for the site relaxed in the δ subunit after its divergence from the α subunit, coinciding with a loss of δ subunit expression in renal tissues. We also show that the proposed Na+ binding site in the α subunit is a bona fide site by conferring novel function to channels comprising human δ subunits. Together, our findings provide evidence that ENaC Na+ self-inhibition improves fitness through its role in Na+ homeostasis in vertebrates.
Collapse
Affiliation(s)
- Xue-Ping Wang
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Priyanka Srinivasan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mustapha El Hamdaoui
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Rafael Grytz
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ossama B Kashlan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Houser A, Baconguis I. Structural Insights into Subunit-Dependent Functional Regulation in Epithelial Sodium Channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.595834. [PMID: 38853903 PMCID: PMC11160588 DOI: 10.1101/2024.05.28.595834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Epithelial sodium channels (ENaC) play a crucial role in Na + reabsorption in mammals. To date, four subunits have been identified-α, β, γ, and δ-believed to form different heteromeric complexes. Currently, only the structure of the αβγ complex is known. To understand how these channels form with varying subunit compositions and define the contribution of each subunit to distinct properties, we co-expressed human δ, β, and γ. Using single-particle cryo-electron microscopy, we observed three distinct ENaC complexes. The structures unveil a pattern in which β and γ positions are conserved among the different complexes while the α position in αβγ trimer is occupied by either δ or another β. The presence of δ induces structural rearrangements in the γ subunit explaining the differences in channel activity observed between αβγ and δβγ channels. These structures define the mechanism by which ENaC subunit composition tunes ENaC function.
Collapse
|
5
|
Zhang J. Hereditary causes of hypertension due to increased sodium transport. Curr Opin Pediatr 2024; 36:211-218. [PMID: 37909881 DOI: 10.1097/mop.0000000000001304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
PURPOSE OF REVIEW Hypertension, commonly known as high blood pressure, is a widespread health condition affecting a large number of individuals across the globe. Although lifestyle choices and environmental factors are known to have a significant impact on its development, there is growing recognition of the influence of genetic factors in the pathogenesis of hypertension. This review specifically focuses on the hereditary causes of hypertension that are associated with increased sodium transport through the thiazide-sensitive NaCl cotransporter (NCC) or amiloride-sensitive epithelial sodium channel (ENaC), crucial mechanisms involved in regulating blood pressure in the kidneys. By examining genetic mutations and signaling molecules linked to the dysregulation of sodium transport, this review aims to deepen our understanding of the hereditary causes of hypertension and shed light on potential therapeutic targets. RECENT FINDINGS Liddle syndrome (LS) is a genetic disorder that typically manifests early in life and is characterized by hypertension, hypokalemic metabolic alkalosis, hyporeninemia, and suppressed aldosterone secretion. This condition is primarily caused by gain-of-function mutations in ENaC. In contrast, Pseudohypoaldosteronism type II (PHAII) is marked by hyperkalemia and hypertension, alongside other clinical features such as hyperchloremia, metabolic acidosis, and suppressed plasma renin levels. PHAII results from overactivations of NCC, brought about by gain-of-function mutations in its upstream signaling molecules, including WNK1 (with no lysine (K) 1), WNK4, Kelch-like 3 (KLHL3), and cullin3 (CUL3). SUMMARY NCC and ENaC are integral components, and their malfunctions lead to disorders like LS and PHAII, hereditary causes of hypertension. Current treatments for LS involve ENaC blockers (e.g., triamterene and amiloride) in conjunction with low-sodium diets, effectively normalizing blood pressure and potassium levels. In PHAII, thiazide diuretics, which inhibit NCC, are the mainstay treatment, albeit with some limitations and potential side effects. Ongoing research in developing alternative treatments, including small molecules targeting key regulators, holds promise for more effective and tailored hypertension solutions.
Collapse
Affiliation(s)
- Jinwei Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Exeter, UK
| |
Collapse
|
6
|
Kashlan OB, Wang XP, Sheng S, Kleyman TR. Epithelial Na + Channels Function as Extracellular Sensors. Compr Physiol 2024; 14:1-41. [PMID: 39109974 PMCID: PMC11309579 DOI: 10.1002/cphy.c230015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
The epithelial Na + channel (ENaC) resides on the apical surfaces of specific epithelia in vertebrates and plays a critical role in extracellular fluid homeostasis. Evidence that ENaC senses the external environment emerged well before the molecular identity of the channel was reported three decades ago. This article discusses progress toward elucidating the mechanisms through which specific external factors regulate ENaC function, highlighting insights gained from structural studies of ENaC and related family members. It also reviews our understanding of the role of ENaC regulation by the extracellular environment in physiology and disease. After familiarizing the reader with the channel's physiological roles and structure, we describe the central role protein allostery plays in ENaC's sensitivity to the external environment. We then discuss each of the extracellular factors that directly regulate the channel: proteases, cations and anions, shear stress, and other regulators specific to particular extracellular compartments. For each regulator, we discuss the initial observations that led to discovery, studies investigating molecular mechanism, and the physiological and pathophysiological implications of regulation. © 2024 American Physiological Society. Compr Physiol 14:5407-5447, 2024.
Collapse
Affiliation(s)
- Ossama B. Kashlan
- Department of Medicine, Renal-Electrolyte Division,
University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xue-Ping Wang
- Department of Medicine, Renal-Electrolyte Division,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaohu Sheng
- Department of Medicine, Renal-Electrolyte Division,
University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas R. Kleyman
- Department of Medicine, Renal-Electrolyte Division,
University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh,
Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
7
|
Liu Y, Xia D, Zhong L, Chen L, Zhang L, Ai M, Mei R, Pang R. Casein Kinase 2 Affects Epilepsy by Regulating Ion Channels: A Potential Mechanism. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:894-905. [PMID: 37350003 DOI: 10.2174/1871527322666230622124618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 06/24/2023]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal brain discharges, is the third most common chronic disorder of the Central Nervous System (CNS). Although significant progress has been made in the research on antiepileptic drugs (AEDs), approximately one-third of patients with epilepsy are refractory to these drugs. Thus, research on the pathogenesis of epilepsy is ongoing to find more effective treatments. Many pathological mechanisms are involved in epilepsy, including neuronal apoptosis, mossy fiber sprouting, neuroinflammation, and dysfunction of neuronal ion channels, leading to abnormal neuronal excitatory networks in the brain. CK2 (Casein kinase 2), which plays a critical role in modulating neuronal excitability and synaptic transmission, has been shown to be associated with epilepsy. However, there is limited research on the mechanisms involved. Recent studies have suggested that CK2 is involved in regulating the function of neuronal ion channels by directly phosphorylating them or their binding partners. Therefore, in this review, we will summarize recent research advances regarding the potential role of CK2 regulating ion channels in epilepsy, aiming to provide more evidence for future studies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Di Xia
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lianmei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ling Chen
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan, 650032, China
| | - Linming Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Mingda Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Rong Mei
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650034, China
| | - Ruijing Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
8
|
Lysikova DV, Vasileva VY, Chubinskiy-Nadezhdin VI, Morachevskaya EA, Sudarikova AV. Capsazepine activates amiloride-insensitive ENaC-like channels in human leukemia cells. Biochem Biophys Res Commun 2023; 687:149187. [PMID: 37944472 DOI: 10.1016/j.bbrc.2023.149187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Sodium influx carried out by ion channels is one of the main regulators of water-salt and volume balance in cells of blood origin. Previously, we described amiloride-insensitive ENaC-like channels in human myeloid leukemia K562 cells; the intracellular regulatory mechanisms of the channels are associated with actin cytoskeleton dynamics. Recently, an extracellular mechanism of ENaC-like channels activation in K562 cells by the action of serine protease trypsin has been revealed. The other extracellular pathways that modulate ENaC (epithelial Na+ channel) activity and sodium permeability in transformed blood cells are not yet fully investigated. Here, we study the action of capsazepine (CPZ), as δ-ENaC activator, on single channel activity in K562 cells in whole-cell patch clamp experiments. Addition of CPZ (2 μM) to the extracellular solution caused an activation of sodium channels with typical features; unitary conductance was 15.1 ± 0.8 pS. Amiloride derivative benzamil (50 μM) did not inhibit their activity. Unitary currents and conductance of CPZ-activated channels were higher in Na+-containing extracellular solution than in Li+, that is one of the main fingerprints of δ-ENaC. The results of RT-PCR analysis and immunofluorescence staining also confirmed the expression of δ-hENaC (as well as α-, β-, γ-ENaC) at the mRNA and protein level. These findings allow us to speculate that CPZ activates amiloride-insensitive ENaC-like channels that contain δ-ENaC in К562 cells. Our data reveal a novel extracellular mechanism for ENaC-like activation in human leukemia cells.
Collapse
Affiliation(s)
- Daria V Lysikova
- Institute of Cytology, Russian Academy of Sciences, 194064 Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Valeria Y Vasileva
- Institute of Cytology, Russian Academy of Sciences, 194064 Tikhoretsky Ave. 4, St. Petersburg, Russia
| | | | - Elena A Morachevskaya
- Institute of Cytology, Russian Academy of Sciences, 194064 Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Anastasia V Sudarikova
- Institute of Cytology, Russian Academy of Sciences, 194064 Tikhoretsky Ave. 4, St. Petersburg, Russia.
| |
Collapse
|
9
|
Chen Y, Yu X, Yan Z, Zhang S, Zhang J, Guo W. Role of epithelial sodium channel-related inflammation in human diseases. Front Immunol 2023; 14:1178410. [PMID: 37559717 PMCID: PMC10407551 DOI: 10.3389/fimmu.2023.1178410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023] Open
Abstract
The epithelial sodium channel (ENaC) is a heterotrimer and is widely distributed throughout the kidneys, blood vessels, lungs, colons, and many other organs. The basic role of the ENaC is to mediate the entry of Na+ into cells; the ENaC also has an important regulatory function in blood pressure, airway surface liquid (ASL), and endothelial cell function. Aldosterone, serum/glucocorticoid kinase 1 (SGK1), shear stress, and posttranslational modifications can regulate the activity of the ENaC; some ion channels also interact with the ENaC. In recent years, it has been found that the ENaC can lead to immune cell activation, endothelial cell dysfunction, aggravated inflammation involved in high salt-induced hypertension, cystic fibrosis, pseudohypoaldosteronism (PHA), and tumors; some inflammatory cytokines have been reported to have a regulatory role on the ENaC. The ENaC hyperfunction mediates the increase of intracellular Na+, and the elevated exchange of Na+ with Ca2+ leads to an intracellular calcium overload, which is an important mechanism for ENaC-related inflammation. Some of the research on the ENaC is controversial or unclear; we therefore reviewed the progress of studies on the role of ENaC-related inflammation in human diseases and their mechanisms.
Collapse
Affiliation(s)
- Yabin Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- National Organ Transplantation (Liver &Kidney Transplantation) Physician Training Centre, Zhengzhou, China
- National Regional Medical Treatment Centre of Henan Organ Transplantation, Zhengzhou, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- National Organ Transplantation (Liver &Kidney Transplantation) Physician Training Centre, Zhengzhou, China
- National Regional Medical Treatment Centre of Henan Organ Transplantation, Zhengzhou, China
| | - Zhiping Yan
- Henan Organ Transplantation Centre, Zhengzhou, China
- Henan Engineering and Research Center for Diagnosis and Treatment of Hepatobiliary and Pancreatic Surgical Diseases, Zhengzhou, China
| | - Shuijun Zhang
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
| | - Jiacheng Zhang
- Henan Key Laboratory for Digestive Organ Transplantation, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory for Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
| |
Collapse
|
10
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
11
|
Abstract
Salt taste, the taste of sodium chloride (NaCl), is mechanistically one of the most complex and puzzling among basic tastes. Sodium has essential functions in the body but causes harm in excess. Thus, animals use salt taste to ingest the right amount of salt, which fluctuates by physiological needs: typically, attraction to low salt concentrations and rejection of high salt. This concentration-valence relationship is universally observed in terrestrial animals, and research has revealed complex peripheral codes for NaCl involving multiple taste pathways of opposing valence. Sodium-dependent and -independent pathways mediate attraction and aversion to NaCl, respectively. Gustatory sensors and cells that transduce NaCl have been uncovered, along with downstream signal transduction and neurotransmission mechanisms. However, much remains unknown. This article reviews classical and recent advances in our understanding of the molecular and cellular mechanisms underlying salt taste in mammals and insects and discusses perspectives on human salt taste.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan; .,Japan Science and Technology Agency, CREST, Saitama, Japan
| | - Michael D Gordon
- Department of Zoology and Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Kasahara Y, Narukawa M, Takeuchi A, Tominaga M, Abe K, Asakura T. Molecular logic of salt taste reception in special reference to transmembrane channel-like 4 (TMC4). J Physiol Sci 2022; 72:31. [PMID: 36451105 PMCID: PMC10717231 DOI: 10.1186/s12576-022-00856-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/13/2022] [Indexed: 12/05/2022]
Abstract
The taste is biologically of intrinsic importance. It almost momentarily perceives environmental stimuli for better survival. In the early 2000s, research into taste reception was greatly developed with discovery of the receptors. However, the mechanism of salt taste reception is not fully elucidated yet and many questions still remain. At present, next-generation sequencing and genome-editing technologies are available which would become pivotal tools to elucidate the remaining issues. Here we review current mechanisms of salt taste reception in particular and characterize the properties of transmembrane channel-like 4 as a novel salt taste-related molecule that we found using these sophisticated tools.
Collapse
Affiliation(s)
- Yoichi Kasahara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-8657, Japan
| | - Masataka Narukawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-8657, Japan
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshicho Imakumano, Higashiyama, Kyoto, 605-8501, Japan
| | - Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, and Life Science Innovation Center, University of Fukui, Fukui, 910-1193, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaijicho, Okazaki, Aichi, 444-8787, Japan
- Thermal Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaijicho, Okazaki, Aichi, 444-8787, Japan
| | - Keiko Abe
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-8657, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), Life Science & Environment Research Center (LiSE), 3-25-13 Tonomachi Kawasaki-Ku, Kawasaki, Kanagawa, 210-0821, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
13
|
Blobner BM, Kirabo A, Kashlan OB, Sheng S, Arnett DK, Becker LC, Boerwinkle E, Carlson JC, Gao Y, Gibbs RA, He J, Irvin MR, Kardia SLR, Kelly TN, Kooperberg C, McGarvey ST, Menon VK, Montasser ME, Naseri T, Redline S, Reiner AP, Reupena MS, Smith JA, Sun X, Vaidya D, Viaud-Martinez KA, Weeks DE, Yanek LR, Zhu X, Minster RL, Kleyman TR. Rare Variants in Genes Encoding Subunits of the Epithelial Na + Channel Are Associated With Blood Pressure and Kidney Function. Hypertension 2022; 79:2573-2582. [PMID: 36193739 PMCID: PMC9669116 DOI: 10.1161/hypertensionaha.121.18513] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 07/31/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The epithelial Na+ channel (ENaC) is intrinsically linked to fluid volume homeostasis and blood pressure. Specific rare mutations in SCNN1A, SCNN1B, and SCNN1G, genes encoding the α, β, and γ subunits of ENaC, respectively, are associated with extreme blood pressure phenotypes. No associations between blood pressure and SCNN1D, which encodes the δ subunit of ENaC, have been reported. A small number of sequence variants in ENaC subunits have been reported to affect functional transport in vitro or blood pressure. The effects of the vast majority of rare and low-frequency ENaC variants on blood pressure are not known. METHODS We explored the association of low frequency and rare variants in the genes encoding ENaC subunits, with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure. Using whole-genome sequencing data from 14 studies participating in the Trans-Omics in Precision Medicine Whole-Genome Sequencing Program, and sequence kernel association tests. RESULTS We found that variants in SCNN1A and SCNN1B were associated with diastolic blood pressure and mean arterial pressure (P<0.00625). Although SCNN1D is poorly expressed in human kidney tissue, SCNN1D variants were associated with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure (P<0.00625). ENaC variants in 2 of the 4 subunits (SCNN1B and SCNN1D) were also associated with estimated glomerular filtration rate (P<0.00625), but not with stroke. CONCLUSIONS Our results suggest that variants in extrarenal ENaCs, in addition to ENaCs expressed in kidneys, influence blood pressure and kidney function.
Collapse
Affiliation(s)
- Brandon M Blobner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ossama B Kashlan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shaohu Sheng
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Lewis C Becker
- GeneSTAR Research Program, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jenna C Carlson
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Gao
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Tulane University Translational Science Institute, New Orleans, LA, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sharon LR Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Tulane University Translational Science Institute, New Orleans, LA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stephen T McGarvey
- Department of Epidemiology and International Health Institute, Brown University School of Public Health, Providence, RI, USA
| | - Vipin K Menon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - May E Montasser
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Take Naseri
- Department of Epidemiology and International Health Institute, Brown University School of Public Health, Providence, RI, USA
- Ministry of Health, Apia, Samoa
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Dhananjay Vaidya
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Daniel E Weeks
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | | | - Ryan L Minster
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Sudarikova AV, Bychkov ML, Kulbatskii DS, Chubinskiy-Nadezhdin VI, Shlepova OV, Shulepko MA, Koshelev SG, Kirpichnikov MP, Lyukmanova EN. Mambalgin-2 Inhibits Lung Adenocarcinoma Growth and Migration by Selective Interaction With ASIC1/α-ENaC/γ-ENaC Heterotrimer. Front Oncol 2022; 12:904742. [PMID: 35837090 PMCID: PMC9273970 DOI: 10.3389/fonc.2022.904742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/24/2022] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is one of the most common cancer types in the world. Despite existing treatment strategies, overall patient survival remains low and new targeted therapies are required. Acidification of the tumor microenvironment drives the growth and metastasis of many cancers. Acid sensors such as acid-sensing ion channels (ASICs) may become promising targets for lung cancer therapy. Previously, we showed that inhibition of the ASIC1 channels by a recombinant analogue of mambalgin-2 from Dendroaspis polylepis controls oncogenic processes in leukemia, glioma, and melanoma cells. Here, we studied the effects and molecular targets of mambalgin-2 in lung adenocarcinoma A549 and Lewis cells, lung transformed WI-38 fibroblasts, and lung normal HLF fibroblasts. We found that mambalgin-2 inhibits the growth and migration of A549, metastatic Lewis P29 cells, and WI-38 cells, but not of normal fibroblasts. A549, Lewis, and WI-38 cells expressed different ASIC and ENaC subunits, while normal fibroblasts did not at all. Mambalgin-2 induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma cells. In line, acidification-evoked inward currents were observed only in A549 and WI-38 cells. Gene knockdown showed that the anti-proliferative and anti-migratory activity of mambalgin-2 is dependent on the expression of ASIC1a, α-ENaC, and γ-ENaC. Using affinity extraction and immunoprecipitation, mambalgin-2 targeting of ASIC1a/α-ENaC/γ-ENaC heteromeric channels in A549 cells was shown. Electrophysiology studies in Xenopus oocytes revealed that mambalgin-2 inhibits the ASIC1a/α-ENaC/γ-ENaC channels with higher efficacy than the ASIC1a channels, pointing on the heteromeric channels as a primary target of the toxin in cancer cells. Finally, bioinformatics analysis showed that the increased expression of ASIC1 and γ-ENaC correlates with a worse survival prognosis for patients with lung adenocarcinoma. Thus, the ASIC1a/α-ENaC/γ-ENaC heterotrimer can be considered a marker of cell oncogenicity and its targeting is promising for the design of new selective cancer therapeutics.
Collapse
Affiliation(s)
- Anastasia V. Sudarikova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Group of Ionic Mechanisms of Cell Signaling, Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Maxim L. Bychkov
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitrii S. Kulbatskii
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vladislav I. Chubinskiy-Nadezhdin
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Group of Ionic Mechanisms of Cell Signaling, Department of Intracellular Signaling and Transport, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga V. Shlepova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Mikhail A. Shulepko
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sergey G. Koshelev
- Laboratory of Neuroreceptors and Neuroregulators, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina N. Lyukmanova
- Laboratory of Bioengineering of Neuromodulators and Neuroreceptors, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Interdisciplinary Scientific and Educational School of Moscow University «Molecular Technologies of the Living Systems and Synthetic Biology», Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- *Correspondence: Ekaterina N. Lyukmanova,
| |
Collapse
|
15
|
Fronius M. Epithelial Na+ channel and the glycocalyx: a sweet and salty relationship for arterial shear stress sensing. Curr Opin Nephrol Hypertens 2022; 31:142-150. [PMID: 34966089 DOI: 10.1097/mnh.0000000000000779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The ability of endothelial cells to sense mechanical force, and shear stress in particular, is crucial for normal vascular function. This relies on an intact endothelial glycocalyx that facilitates the production of nitric oxide (NO). An emerging arterial shear stress sensor is the epithelial Na+ channel (ENaC). This review highlights existing and new evidence for the interdependent activity of the glycocalyx and ENaC and its implications for vascular function. RECENT FINDINGS New evidence suggests that the glycocalyx and ENaC are physically connected and that this is important for shear stress sensing. The connection relies on N-glycans attached to glycosylated asparagines of α-ENaC. Removal of specific N-glycans reduced ENaC's shear stress response. Similar effects were observed following degradation of the glycocalyx. Endothelial specific viral transduction of α-ENaC increased blood pressure (∼40 mmHg). This increase was attenuated in animals transduced with an α-ENaC version lacking N-glycans. SUMMARY These observations indicate that ENaC is connected to the glycocalyx and their activity is interdependent to facilitate arterial shear stress sensation. Future research focusing on how N-glycans mediate this interaction can provide new insights for the understanding of vascular function in health and disease.
Collapse
Affiliation(s)
- Martin Fronius
- Department of Physiology, School of Biomedical Sciences
- HeartOtago, University of Otago, Dunedin
- Healthy Hearts Aotearoa New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Centre of Research Excellence, Auckland, New Zealand
| |
Collapse
|
16
|
Wang XP, Balchak DM, Gentilcore C, Clark NL, Kashlan OB. Activation by cleavage of the epithelial Na + channel α and γ subunits independently coevolved with the vertebrate terrestrial migration. eLife 2022; 11:75796. [PMID: 34984981 PMCID: PMC8791634 DOI: 10.7554/elife.75796] [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: 11/23/2021] [Accepted: 12/28/2021] [Indexed: 12/17/2022] Open
Abstract
Vertebrates evolved mechanisms for sodium conservation and gas exchange in conjunction with migration from aquatic to terrestrial habitats. Epithelial Na+ channel (ENaC) function is critical to systems responsible for extracellular fluid homeostasis and gas exchange. ENaC is activated by cleavage at multiple specific extracellular polybasic sites, releasing inhibitory tracts from the channel’s α and γ subunits. We found that proximal and distal polybasic tracts in ENaC subunits coevolved, consistent with the dual cleavage requirement for activation observed in mammals. Polybasic tract pairs evolved with the terrestrial migration and the appearance of lungs, coincident with the ENaC activator aldosterone, and appeared independently in the α and γ subunits. In summary, sites within ENaC for protease activation developed in vertebrates when renal Na+ conservation and alveolar gas exchange were required for terrestrial survival.
Collapse
Affiliation(s)
- Xue-Ping Wang
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Deidra M Balchak
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Clayton Gentilcore
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Nathan L Clark
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Ossama B Kashlan
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States.,Computational and Systems Biology, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
17
|
Yan Y, Wang J, Yu L, Cui B, Wang H, Xiao X, Zhang Y, Zheng J, Wang J, Hui R, Wang Y. ANKRD36 Is Involved in Hypertension by Altering Expression of ENaC Genes. Circ Res 2021; 129:1067-1081. [PMID: 34615377 DOI: 10.1161/circresaha.121.319883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yupeng Yan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Jin'e Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Liang Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Hongrui Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Xiao Xiao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Yu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Jun Zheng
- Rizhao Port Hospital, Shandong, China (J.Z., Jingjun Wang)
| | - Jingjun Wang
- Rizhao Port Hospital, Shandong, China (J.Z., Jingjun Wang)
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.Y., Jin'e Wang, L.Y., B.C., H.W., X.X., Y.Z., R.H., Y.W.)
| |
Collapse
|
18
|
Gettings SM, Maxeiner S, Tzika M, Cobain MRD, Ruf I, Benseler F, Brose N, Krasteva-Christ G, Vande Velde G, Schönberger M, Althaus M. Two functional epithelial sodium channel isoforms are present in rodents despite pronounced evolutionary pseudogenisation and exon fusion. Mol Biol Evol 2021; 38:5704-5725. [PMID: 34491346 PMCID: PMC8662647 DOI: 10.1093/molbev/msab271] [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] [Indexed: 12/30/2022] Open
Abstract
The epithelial sodium channel (ENaC) plays a key role in salt and water homeostasis in
tetrapod vertebrates. There are four ENaC subunits (α, β, γ, δ), forming heterotrimeric
αβγ- or δβγ-ENaCs. Although the physiology of αβγ-ENaC is well understood, for decades the
field has stalled with respect to δβγ-ENaC due to the lack of mammalian model organisms.
The SCNN1D gene coding for δ-ENaC was previously believed to be absent in
rodents, hindering studies using standard laboratory animals. We analyzed all currently
available rodent genomes and discovered that SCNN1D is present in rodents
but was independently lost in five rodent lineages, including the Muridae (mice and rats).
The independent loss of SCNN1D in rodent lineages may be constrained by
phylogeny and taxon-specific adaptation to dry habitats, however habitat aridity does not
provide a selection pressure for maintenance of SCNN1D across Rodentia. A
fusion of two exons coding for a structurally flexible region in the extracellular domain
of δ-ENaC appeared in the Hystricognathi (a group that includes guinea pigs). This
conserved pattern evolved at least 41 Ma and represents a new autapomorphic feature for
this clade. Exon fusion does not impair functionality of guinea pig (Cavia
porcellus) δβγ-ENaC expressed in Xenopus oocytes.
Electrophysiological characterization at the whole-cell and single-channel level revealed
conserved biophysical features and mechanisms controlling guinea pig αβγ- and δβγ-ENaC
function as compared with human orthologs. Guinea pigs therefore represent commercially
available mammalian model animals that will help shed light on the physiological function
of δ-ENaC.
Collapse
Affiliation(s)
- Sean M Gettings
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Stephan Maxeiner
- Institute for Anatomy and Cell Biology, Saarland University School of Medicine, Homburg, Germany
| | - Maria Tzika
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthew R D Cobain
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Irina Ruf
- Division of Messel Research and Mammalogy, Senckenberg Research Institute and Natural History Museum Frankfurt, Frankfurt am Main, Germany
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Gabriela Krasteva-Christ
- Institute for Anatomy and Cell Biology, Saarland University School of Medicine, Homburg, Germany
| | - Greetje Vande Velde
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Matthias Schönberger
- Biomedical Imaging, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Belgium
| | - Mike Althaus
- Institute for Functional Gene Analytics, Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| |
Collapse
|
19
|
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
|
20
|
Zhu Z, Mei J, Sun S, Lu S, Li M, Guan Y, Chen Y, Xu Y, Zhang T, Shi F, Li X, Miao M, Zhao S, Gao Q, Mi Q, Tang P, Yao J. Nutrigenomics reveals potential genetic underpinning of diverse taste preference of Chinese men. Genes Genomics 2021; 43:689-699. [PMID: 33843022 DOI: 10.1007/s13258-021-01079-y] [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: 10/27/2020] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Taste preference varies geographically in China. However, studies on Chinese people's taste preference in different regions of China are limited, and are lack of research on the mechanism of differences in taste preference, especially in genetics. OBJECTIVE This study aims to investigate the characteristics of taste preference of Chinese men, and estimate whether diverse taste preference in Chinese have genetic underpinning. METHODS We conducted a questionnaire survey on taste preferences on 1076 males from 10 regions of China, and collected another 1427 males from the same regions which genotyped by microarray. We compared the correlation between different taste preference, and evaluated the correlation between the mutation frequency of inhouse database and different taste preference. The putative taste-preference-related genes were further utilized to estimate the candidate relationship on gene and gene network in different taste preference. RESULTS There was a correlation between different taste preferences in Chinese men. We found 31 SNPs associated with 6 kind of taste preferences. These SNPs located within or nearby 36 genes, and the tastes associated with 4 of these genes (TRPV1, AGT, ASIC2 and GLP1R) are consistent with the previous studies. Moreover, in different tastes which were suggested to be associated with each other, some putative related genes were the same or in the same gene network, such as pathways related with blood pressure, response to stimulus and nervous system. CONCLUSIONS This study indicates that the diverse taste preference of Chinese men may have genetic underpinning.
Collapse
Affiliation(s)
- Zhouhai Zhu
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Junpu Mei
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Silong Sun
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Sheming Lu
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Meng Li
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Ying Guan
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Ying Chen
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Yuqiong Xu
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Tao Zhang
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Fengxue Shi
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Xuemei Li
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Mingming Miao
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Shancen Zhao
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Qian Gao
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Qili Mi
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Ping Tang
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China
| | - Jianhua Yao
- Department of Tobacco and Health, Joint Institute of Tobacco and Health, 41 Keyi Road, Wuhua District, Kunming, 650106, Yunnan, China.
| |
Collapse
|
21
|
Abstract
The Epithelial Na+ Channel, ENaC, comprised of 3 subunits (αβγ, or sometimes δβγENaC), plays a critical role in regulating salt and fluid homeostasis in the body. It regulates fluid reabsorption into the blood stream from the kidney to control blood volume and pressure, fluid absorption in the lung to control alveolar fluid clearance at birth and maintenance of normal airway surface liquid throughout life, and fluid absorption in the distal colon and other epithelial tissues. Moreover, recent studies have also revealed a role for sodium movement via ENaC in nonepithelial cells/tissues, such as endothelial cells in blood vessels and neurons. Over the past 25 years, major advances have been made in our understanding of ENaC structure, function, regulation, and role in human disease. These include the recently solved three-dimensional structure of ENaC, ENaC function in various tissues, and mutations in ENaC that cause a hereditary form of hypertension (Liddle syndrome), salt-wasting hypotension (PHA1), or polymorphism in ENaC that contributes to other diseases (such as cystic fibrosis). Moreover, great strides have been made in deciphering the regulation of ENaC by hormones (e.g., the mineralocorticoid aldosterone, glucocorticoids, vasopressin), ions (e.g., Na+ ), proteins (e.g., the ubiquitin-protein ligase NEDD4-2, the kinases SGK1, AKT, AMPK, WNKs & mTORC2, and proteases), and posttranslational modifications [e.g., (de)ubiquitylation, glycosylation, phosphorylation, acetylation, palmitoylation]. Characterization of ENaC structure, function, regulation, and role in human disease, including using animal models, are described in this article, with a special emphasis on recent advances in the field. © 2021 American Physiological Society. Compr Physiol 11:1-29, 2021.
Collapse
Affiliation(s)
- Daniela Rotin
- The Hospital for Sick Children, and The University of Toronto, Toronto, Canada
| | - Olivier Staub
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
22
|
Lossow K, Hermans-Borgmeyer I, Meyerhof W, Behrens M. Segregated Expression of ENaC Subunits in Taste Cells. Chem Senses 2021; 45:235-248. [PMID: 32006019 DOI: 10.1093/chemse/bjaa004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Salt taste is one of the 5 basic taste qualities. Depending on the concentration, table salt is perceived either as appetitive or aversive, suggesting the contribution of several mechanisms to salt taste, distinguishable by their sensitivity to the epithelial sodium channel (ENaC) blocker amiloride. A taste-specific knockout of the α-subunit of the ENaC revealed the relevance of this polypeptide for low-salt transduction, whereas the response to other taste qualities remained normal. The fully functional ENaC is composed of α-, β-, and γ-subunits. In taste tissue, however, the precise constitution of the channel and the cell population responsible for detecting table salt remain uncertain. In order to examine the cells and subunits building the ENaC, we generated mice carrying modified alleles allowing the synthesis of green and red fluorescent proteins in cells expressing the α- and β-subunit, respectively. Fluorescence signals were detected in all types of taste papillae and in taste buds of the soft palate and naso-incisor duct. However, the lingual expression patterns of the reporters differed depending on tongue topography. Additionally, immunohistochemistry for the γ-subunit of the ENaC revealed a lack of overlap between all potential subunits. The data suggest that amiloride-sensitive recognition of table salt is unlikely to depend on the classical ENaCs formed by α-, β-, and γ-subunits and ask for a careful investigation of the channel composition.
Collapse
Affiliation(s)
- Kristina Lossow
- Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee, Nuthetal, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animal Unit, University Medical Center Hamburg-Eppendorf (ZMNH), Hamburg, Germany
| | - Wolfgang Meyerhof
- Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee, Nuthetal, Germany
| | - Maik Behrens
- Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee, Nuthetal, Germany
| |
Collapse
|
23
|
Abstract
Among the 5 taste qualities, salt is the least understood. The receptors, their expression pattern in taste cells, and the transduction mechanisms for salt taste are still unclear. Previous studies have suggested that low concentrations of NaCl are detected by the amiloride-sensitive epithelial Na+ channel (ENaC), which in other systems requires assembly of 3 homologous subunits (α, β, and γ) to form a functional channel. However, a new study from Lossow and colleagues, published in this issue of Chemical Senses, challenges that hypothesis by examining expression levels of the 3 ENaC subunits in individual taste cells using gene-targeted mice in combination with immunohistochemistry and in situ hybridization. Results show a lack of colocalization of ENaC subunits in taste cells as well as expression of subunits in taste cells that show no amiloride sensitivity. These new results question the molecular identity of the amiloride-sensitive Na+ conductance in taste cells.
Collapse
Affiliation(s)
- Aurelie Vandenbeuch
- Department of Otolaryngology and Rocky Mountain Taste and Smell Center, University of Colorado-Anschutz Medical Campus, Aurora, USA
| | - Sue C Kinnamon
- Department of Otolaryngology and Rocky Mountain Taste and Smell Center, University of Colorado-Anschutz Medical Campus, Aurora, USA
| |
Collapse
|
24
|
Abstract
The development of high blood pressure is influenced by genetic and environmental factors, with high salt intake being a known environmental contributor. Humans display a spectrum of sodium-sensitivity, with some individuals displaying a significant blood pressure rise in response to increased sodium intake while others experience almost no change. These differences are, in part, attributable to genetic variation in pathways involved in sodium handling and excretion. ENaC (epithelial sodium channel) is one of the key transporters responsible for the reabsorption of sodium in the distal nephron. This channel has an important role in the regulation of extracellular fluid volume and consequently blood pressure. Herein, we review the role of ENaC in the development of salt-sensitive hypertension, and present mechanistic insights into the regulation of ENaC activity and how it may accelerate sodium-induced damage and dysfunction. We discuss the traditional role of ENaC in renal sodium reabsorption and review work addressing ENaC expression and function in the brain, vasculature, and immune cells, and how this has expanded the implications for its role in the initiation and progression of salt-sensitive hypertension.
Collapse
Affiliation(s)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, and Department of Molecular Physiology and Biophysics Vanderbilt University, Nashville, TN (A.K.)
| | - Thomas R Kleyman
- From the Department of Medicine (S.M.M., T.R.K.), University of Pittsburgh, PA.,Department of Cell Biology (T.R.K.), University of Pittsburgh, PA.,Department of Pharmacology and Chemical Biology (T.R.K.), University of Pittsburgh, PA
| |
Collapse
|
25
|
Paudel P, McDonald FJ, Fronius M. The δ subunit of epithelial sodium channel in humans-a potential player in vascular physiology. Am J Physiol Heart Circ Physiol 2020; 320:H487-H493. [PMID: 33275523 DOI: 10.1152/ajpheart.00800.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular epithelial sodium channels (ENaCs) made up of canonical α, β, and γ subunits have attracted more attention recently owing to their physiological role in vascular health and disease. A fourth subunit, δ-ENaC, is expressed in various mammalian species, except mice and rats, which are common animal models for cardiovascular research. Accordingly, δ-ENaC is the least understood subunit. However, the recent discovery of δ subunit in human vascular cells indicates that this subunit may play a significant role in normal/pathological vascular physiology in humans. Channels containing the δ subunit have different biophysical and pharmacological properties compared with channels containing the α subunit, with the potential to alter the vascular function of ENaC in health and disease. Hence, it is important to investigate the expression and function of δ-ENaC in the vasculature to identify whether δ-ENaC is a potential new drug target for the treatment of cardiovascular disease. In this review, we will focus on the existing knowledge of δ-ENaC and implications for vascular physiology and pathophysiology in humans.
Collapse
Affiliation(s)
- Puja Paudel
- Department of Physiology, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Fiona J McDonald
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
26
|
Montenarh M, Götz C. Protein kinase CK2 and ion channels (Review). Biomed Rep 2020; 13:55. [PMID: 33082952 PMCID: PMC7560519 DOI: 10.3892/br.2020.1362] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Protein kinase CK2 appears as a tetramer or higher molecular weight oligomer composed of catalytic CK2α, CK2α' subunits and non-catalytic regulatory CK2β subunits or as individual subunits. It is implicated in a variety of different regulatory processes, such as Akt signalling, splicing and DNA repair within eukaryotic cells. The present review evaluates the influence of CK2 on ion channels in the plasma membrane. CK2 phosphorylates platform proteins such as calmodulin and ankyrin G, which bind to channel proteins for a physiological transport to and positioning into the membrane. In addition, CK2 directly phosphorylates a variety of channel proteins directly to regulate opening and closing of the channels. Thus, modulation of CK2 activities by specific inhibitors, by siRNA technology or by CRISPR/Cas technology has an influence on intracellular ion concentrations and thereby on cellular signalling. The physiological regulation of the intracellular ion concentration is important for cell survival and correct intracellular signalling. Disturbance of this regulation results in a variety of different diseases including epilepsy, heart failure, cystic fibrosis and diabetes. Therefore, these effects should be considered when using CK2 inhibition as a treatment option for cancer.
Collapse
Affiliation(s)
- Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| |
Collapse
|
27
|
Brecker M, Khakhina S, Schubert TJ, Thompson Z, Rubenstein RC. The Probable, Possible, and Novel Functions of ERp29. Front Physiol 2020; 11:574339. [PMID: 33013490 PMCID: PMC7506106 DOI: 10.3389/fphys.2020.574339] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022] Open
Abstract
The luminal endoplasmic reticulum (ER) protein of 29 kDa (ERp29) is a ubiquitously expressed cellular agent with multiple critical roles. ERp29 regulates the biosynthesis and trafficking of several transmembrane and secretory proteins, including the cystic fibrosis transmembrane conductance regulator (CFTR), the epithelial sodium channel (ENaC), thyroglobulin, connexin 43 hemichannels, and proinsulin. ERp29 is hypothesized to promote ER to cis-Golgi cargo protein transport via COP II machinery through its interactions with the KDEL receptor; this interaction may facilitate the loading of ERp29 clients into COP II vesicles. ERp29 also plays a role in ER stress (ERS) and the unfolded protein response (UPR) and is implicated in oncogenesis. Here, we review the vast array of ERp29’s clients, its role as an ER to Golgi escort protein, and further suggest ERp29 as a potential target for therapies related to diseases of protein misfolding and mistrafficking.
Collapse
Affiliation(s)
- Margaret Brecker
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Svetlana Khakhina
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tyler J. Schubert
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Zachary Thompson
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ronald C. Rubenstein
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- *Correspondence: Ronald C. Rubenstein, ;
| |
Collapse
|
28
|
Wichmann L, Althaus M. Evolution of epithelial sodium channels: current concepts and hypotheses. Am J Physiol Regul Integr Comp Physiol 2020; 319:R387-R400. [PMID: 32783689 DOI: 10.1152/ajpregu.00144.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The conquest of freshwater and terrestrial habitats was a key event during vertebrate evolution. Occupation of low-salinity and dry environments required significant osmoregulatory adaptations enabling stable ion and water homeostasis. Sodium is one of the most important ions within the extracellular liquid of vertebrates, and molecular machinery for urinary reabsorption of this electrolyte is critical for the maintenance of body osmoregulation. Key ion channels involved in the fine-tuning of sodium homeostasis in tetrapod vertebrates are epithelial sodium channels (ENaCs), which allow the selective influx of sodium ions across the apical membrane of epithelial cells lining the distal nephron or the colon. Furthermore, ENaC-mediated sodium absorption across tetrapod lung epithelia is crucial for the control of liquid volumes lining the pulmonary surfaces. ENaCs are vertebrate-specific members of the degenerin/ENaC family of cation channels; however, there is limited knowledge on the evolution of ENaC within this ion channel family. This review outlines current concepts and hypotheses on ENaC phylogeny and discusses the emergence of regulation-defining sequence motifs in the context of osmoregulatory adaptations during tetrapod terrestrialization. In light of the distinct regulation and expression of ENaC isoforms in tetrapod vertebrates, we discuss the potential significance of ENaC orthologs in osmoregulation of fishes as well as the putative fates of atypical channel isoforms in mammals. We hypothesize that ancestral proton-sensitive ENaC orthologs might have aided the osmoregulatory adaptation to freshwater environments whereas channel regulation by proteases evolved as a molecular adaptation to lung liquid homeostasis in terrestrial tetrapods.
Collapse
Affiliation(s)
- Lukas Wichmann
- Institute for Animal Physiology, Justus Liebig University, Giessen, Germany
| | - Mike Althaus
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| |
Collapse
|
29
|
Baldin JP, Barth D, Fronius M. Epithelial Na + Channel (ENaC) Formed by One or Two Subunits Forms Functional Channels That Respond to Shear Force. Front Physiol 2020; 11:141. [PMID: 32256376 PMCID: PMC7090232 DOI: 10.3389/fphys.2020.00141] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
Canonical epithelial sodium channels (ENaCs) are heterotrimers formed by α, β, and γ ENaC subunits in vertebrates and belong to the Degenerin/ENaC family of proteins. Proteins from this family form mechanosensitive channels throughout the animal kingdom. Activity of canonical ENaC is regulated by shear force (SF) mediating Na+ absorption in the kidney and vascular tone of arteries. Expression analysis suggests that non-canonical ENaC, formed by single or only two subunits, exist in certain tissues, but it is unknown if these channels respond to SF. α, β, γ, and δ ENaC subunits were expressed either alone or in combinations of two subunits in Xenopus oocytes. Amiloride-sensitive currents and the responses to SF were assessed using two-electrode voltage clamp recordings. With the exception of γ ENaC, all homomeric channels provided amiloride-sensitive currents and responded to SF applied via a fluid stream directed onto the oocytes. Channels containing two subunits were also activated by SF. Here, the presence of the γ ENaC subunit when co-expressed with α or δ augmented the SF response in comparison to the αβγ/δβγ ENaC. Overall, we provide evidence that non-canonical ENaC can form channels that respond to SF. This supports a potential function of non-canonical ENaC as mechanosensors in epithelial, vascular, and sensory cells.
Collapse
Affiliation(s)
- Jan-Peter Baldin
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Daniel Barth
- Institute of Physiology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Martin Fronius
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
30
|
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
|
31
|
Li Q, Fung E. Multifaceted Functions of Epithelial Na + Channel in Modulating Blood Pressure. Hypertension 2019; 73:273-281. [PMID: 30580685 DOI: 10.1161/hypertensionaha.118.12330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qi Li
- From the Division of Cardiology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (Q.L., E.F.).,Laboratory for Heart Failure and Circulation Research, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Hong Kong SAR (Q.L., E.F.)
| | - Erik Fung
- From the Division of Cardiology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (Q.L., E.F.).,Gerald Choa Cardiac Research Centre, Faculty of Medicine, The Chinese University of Hong Kong (E.F.).,Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong (E.F.).,Laboratory for Heart Failure and Circulation Research, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Hong Kong SAR (Q.L., E.F.)
| |
Collapse
|
32
|
Zhao R, Ali G, Chang J, Komatsu S, Tsukasaki Y, Nie HG, Chang Y, Zhang M, Liu Y, Jain K, Jung BG, Samten B, Jiang D, Liang J, Ikebe M, Matthay MA, Ji HL. Proliferative regulation of alveolar epithelial type 2 progenitor cells by human Scnn1d gene. Am J Cancer Res 2019; 9:8155-8170. [PMID: 31754387 PMCID: PMC6857051 DOI: 10.7150/thno.37023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/16/2019] [Indexed: 01/03/2023] Open
Abstract
Lung epithelial sodium channel (ENaC) encoded by Scnn1 genes is essential for maintaining transepithelial salt and fluid homeostasis in the airway and the lung. Compared to α, β, and γ subunits, the role of respiratory δ-ENaC has not been studied in vivo due to the lack of animal models. Methods: We characterized full-length human δ802-ENaC expressed in both Xenopus oocytes and humanized transgenic mice. AT2 proliferation and differentiation in 3D organoids were analysed with FACS and a confocal microscope. Both two-electrode voltage clamp and Ussing chamber systems were applied to digitize δ802-ENaC channel activity. Immunoblotting was utilized to analyse δ802-ENaC protein. Transcripts of individual ENaC subunits in human lung tissues were quantitated with qPCR. Results: The results indicate that δ802-ENaC functions as an amiloride-inhibitable Na+ channel. Inhibitory peptide α-13 distinguishes δ802- from α-type ENaC channels. Modified proteolysis of γ-ENaC by plasmin and aprotinin did not alter the inhibition of amiloride and α-13 peptide. Expression of δ802-ENaC at the apical membrane of respiratory epithelium was detected with biophysical features similar to those of heterologously expressed channels in oocytes. δ802-ENaC regulated alveologenesis through facilitating the proliferation of alveolar type 2 epithelial cells. Conclusion: The humanized mouse line conditionally expressing human δ802-ENaC is a novel model for studying the expression and function of this protein in vivo .
Collapse
|
33
|
Wichmann L, Dulai JS, Marles-Wright J, Maxeiner S, Szczesniak PP, Manzini I, Althaus M. An extracellular acidic cleft confers profound H +-sensitivity to epithelial sodium channels containing the δ-subunit in Xenopus laevis. J Biol Chem 2019; 294:12507-12520. [PMID: 31248986 DOI: 10.1074/jbc.ra119.008255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/27/2019] [Indexed: 11/06/2022] Open
Abstract
The limited sodium availability of freshwater and terrestrial environments was a major physiological challenge during vertebrate evolution. The epithelial sodium channel (ENaC) is present in the apical membrane of sodium-absorbing vertebrate epithelia and evolved as part of a machinery for efficient sodium conservation. ENaC belongs to the degenerin/ENaC protein family and is the only member that opens without an external stimulus. We hypothesized that ENaC evolved from a proton-activated sodium channel present in ionocytes of freshwater vertebrates and therefore investigated whether such ancestral traits are present in ENaC isoforms of the aquatic pipid frog Xenopus laevis Using whole-cell and single-channel electrophysiology of Xenopus oocytes expressing ENaC isoforms assembled from αβγ- or δβγ-subunit combinations, we demonstrate that Xenopus δβγ-ENaC is profoundly activated by extracellular acidification within biologically relevant ranges (pH 8.0-6.0). This effect was not observed in Xenopus αβγ-ENaC or human ENaC orthologs. We show that protons interfere with allosteric ENaC inhibition by extracellular sodium ions, thereby increasing the probability of channel opening. Using homology modeling of ENaC structure and site-directed mutagenesis, we identified a cleft region within the extracellular loop of the δ-subunit that contains several acidic amino acid residues that confer proton-sensitivity and enable allosteric inhibition by extracellular sodium ions. We propose that Xenopus δβγ-ENaC can serve as a model for investigating ENaC transformation from a proton-activated toward a constitutively-active ion channel. Such transformation might have occurred during the evolution of tetrapod vertebrates to enable bulk sodium absorption during the water-to-land transition.
Collapse
Affiliation(s)
- Lukas Wichmann
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, United Kingdom; Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University Giessen, 35390 Giessen, Germany
| | - Jasdip Singh Dulai
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Jon Marles-Wright
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Pawel Piotr Szczesniak
- Department of Medicine, Haematology/Oncology, Johann-Wolfgang-Goethe University Frankfurt, 60323 Frankfurt, Germany
| | - Ivan Manzini
- Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University Giessen, 35390 Giessen, Germany
| | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, United Kingdom.
| |
Collapse
|
34
|
Dobon B, Rossell C, Walsh S, Bertranpetit J. Is there adaptation in the human genome for taste perception and phase I biotransformation? BMC Evol Biol 2019; 19:39. [PMID: 30704392 PMCID: PMC6357387 DOI: 10.1186/s12862-019-1366-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022] Open
Abstract
Background During the modern human expansion, new environmental pressures may have driven adaptation, especially in genes related to the perception of ingested substances and their detoxification. Consequently, positive (adaptive) selection may have occurred in genes related to taste, and in those related to the CYP450 system due to its role in biotransformation of potentially toxic compounds. A total of 91 genes (taste receptors and CYP450 superfamily) have been studied using Hierarchical Boosting, a powerful combination of different selection tests, to detect signatures of recent positive selection in three continental human populations: Northern Europeans (CEU), East Asians (CHB) and Africans (YRI). Analyses have been refined with selection analyses of the 26 populations of 1000 Genomes Project Phase 3. Results Genes related to taste perception have not been positively selected in the three continental human populations. This finding suggests that, contrary to results of previous studies, different allele frequencies among populations in genes such as TAS2R38 and TAS2R16 are not due to positive selection but to genetic drift. CYP1 and CYP2 genes, also previously considered to be under positive selection, did not show signatures of selective sweeps. However, three genes belonging to the CYP450 system have been identified by the Hierarchical Boosting as positively selected: CYP3A4 and CYP3A43 in CEU, and CYP27A1 in CHB. Conclusions No main adaptive differences are found in known taste receptor genes among the three continental human populations studied. However, there are important genetic adaptations in the cytochrome P450 system related to the Out of Africa expansion of modern humans. We confirmed that CYP3A4 and CYP3A43 are under selection in CEU, and we report for the first time CYP27A1 to be under positive selection in CHB. Electronic supplementary material The online version of this article (10.1186/s12862-019-1366-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Begoña Dobon
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88. 08003, Barcelona, Catalonia, Spain
| | - Carla Rossell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Tomtebodavägen 23a, 17165, Stockholm, Solna, Sweden
| | - Sandra Walsh
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88. 08003, Barcelona, Catalonia, Spain
| | - Jaume Bertranpetit
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88. 08003, Barcelona, Catalonia, Spain.
| |
Collapse
|
35
|
Kandel C, Schmidt P, Perniss A, Keshavarz M, Scholz P, Osterloh S, Althaus M, Kummer W, Deckmann K. ENaC in Cholinergic Brush Cells. Front Cell Dev Biol 2018; 6:89. [PMID: 30159312 PMCID: PMC6103785 DOI: 10.3389/fcell.2018.00089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022] Open
Abstract
Cholinergic polymodal chemosensory cells in the mammalian urethra (urethral brush cells = UBC) functionally express the canonical bitter and umami taste transduction signaling cascade. Here, we aimed to determine whether UBC are functionally equipped for the perception of salt through ENaC (epithelial sodium channel). Cholinergic UBC were isolated from ChAT-eGFP reporter mice (ChAT = choline acetyltransferase). RT-PCR showed mRNA expression of ENaC subunits Scnn1a, Scnn1b, and Scnn1g in urethral epithelium and isolated UBC. Scnn1a could also be detected by next generation sequencing in 4/6 (66%) single UBC, two of them also expressed the bitter receptor Tas2R108. Strong expression of Scnn1a was seen in some urothelial umbrella cells and in 65% of UBC (30/46 cells) in a Scnn1a reporter mouse strain. Intracellular [Ca2+] was recorded in isolated UBC stimulated with the bitter substance denatonium benzoate (25 mM), ATP (0.5 mM) and NaCl (50 mM, on top of 145 mM Na+ and 153 mM Cl− baseline in buffer); mannitol (150 mM) served as osmolarity control. NaCl, but not mannitol, evoked an increase in intracellular [Ca2+] in 70% of the tested UBC. The NaCl-induced effect was blocked by the ENaC inhibitor amiloride (IC50 = 0.47 μM). When responses to both NaCl and denatonium were tested, all three possible positive response patterns occurred in a balanced distribution: 42% NaCl only, 33% denatonium only, 25% to both stimuli. A similar reaction pattern was observed with ATP and NaCl as test stimuli. About 22% of the UBC reacted to all three stimuli. Thus, NaCl evokes calcium responses in several UBC, likely involving an amiloride-sensitive channel containing α-ENaC. This feature does not define a new subpopulation of UBC, but rather emphasizes their polymodal character. The actual function of α-ENaC in cholinergic UBC—salt perception, homeostatic ion transport, mechanoreception—remains to be determined.
Collapse
Affiliation(s)
- Chrissy Kandel
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Patricia Schmidt
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Alexander Perniss
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Maryam Keshavarz
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Paul Scholz
- Department of Cell Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Sabrina Osterloh
- Department of Cell Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Wolfgang Kummer
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Klaus Deckmann
- Institute for Anatomy and Cell Biology, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
36
|
Tagalakis AD, Munye MM, Ivanova R, Chen H, Smith CM, Aldossary AM, Rosa LZ, Moulding D, Barnes JL, Kafetzis KN, Jones SA, Baines DL, Moss GWJ, O'Callaghan C, McAnulty RJ, Hart SL. Effective silencing of ENaC by siRNA delivered with epithelial-targeted nanocomplexes in human cystic fibrosis cells and in mouse lung. Thorax 2018; 73:847-856. [PMID: 29748250 PMCID: PMC6109249 DOI: 10.1136/thoraxjnl-2017-210670] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Loss of the cystic fibrosis transmembrane conductance regulator in cystic fibrosis (CF) leads to hyperabsorption of sodium and fluid from the airway due to upregulation of the epithelial sodium channel (ENaC). Thickened mucus and depleted airway surface liquid (ASL) then lead to impaired mucociliary clearance. ENaC regulation is thus a promising target for CF therapy. Our aim was to develop siRNA nanocomplexes that mediate effective silencing of airway epithelial ENaC in vitro and in vivo with functional correction of epithelial ion and fluid transport. METHODS We investigated translocation of nanocomplexes through mucus and their transfection efficiency in primary CF epithelial cells grown at air-liquid interface (ALI).Short interfering RNA (SiRNA)-mediated silencing was examined by quantitative RT-PCR and western analysis of ENaC. Transepithelial potential (Vt), short circuit current (Isc), ASL depth and ciliary beat frequency (CBF) were measured for functional analysis. Inflammation was analysed by histological analysis of normal mouse lung tissue sections. RESULTS Nanocomplexes translocated more rapidly than siRNA alone through mucus. Transfections of primary CF epithelial cells with nanocomplexes targeting αENaC siRNA, reduced αENaC and βENaC mRNA by 30%. Transfections reduced Vt, the amiloride-sensitive Isc and mucus protein concentration while increasing ASL depth and CBF to normal levels. A single dose of siRNA in mouse lung silenced ENaC by approximately 30%, which persisted for at least 7 days. Three doses of siRNA increased silencing to approximately 50%. CONCLUSION Nanoparticle-mediated delivery of ENaCsiRNA to ALI cultures corrected aspects of the mucociliary defect in human CF cells and offers effective delivery and silencing in vivo.
Collapse
Affiliation(s)
- Aristides D Tagalakis
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Mustafa M Munye
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rositsa Ivanova
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Hanpeng Chen
- Institute of Pharmaceutical Science, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Claire M Smith
- Respiratory, Critical Care and Anaesthesia, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ahmad M Aldossary
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Luca Z Rosa
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dale Moulding
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Konstantinos N Kafetzis
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Stuart A Jones
- Institute of Pharmaceutical Science, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Deborah L Baines
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Guy W J Moss
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Christopher O'Callaghan
- Respiratory, Critical Care and Anaesthesia, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robin J McAnulty
- UCL Respiratory Centre for Inflammation and Tissue Repair, London, UK
| | - Stephen L Hart
- Experimental and Personalised Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
37
|
Wichmann L, Vowinkel KS, Perniss A, Manzini I, Althaus M. Incorporation of the δ-subunit into the epithelial sodium channel (ENaC) generates protease-resistant ENaCs in Xenopus laevis. J Biol Chem 2018; 293:6647-6658. [PMID: 29576549 DOI: 10.1074/jbc.ra118.002543] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/14/2018] [Indexed: 11/06/2022] Open
Abstract
The epithelial sodium channel (ENaC) is a critical regulator of vertebrate electrolyte homeostasis. ENaC is the only constitutively open ion channel in the degenerin/ENaC protein family, and its expression, membrane abundance, and open probability therefore are tightly controlled. The canonical ENaC is composed of three subunits (α, β, and γ), but a fourth δ-subunit may replace α and form atypical δβγ-ENaCs. Using Xenopus laevis as a model, here we found that mRNAs of the α- and δ-subunits are differentially expressed in different tissues and that δ-ENaC predominantly is present in the urogenital tract. Using whole-cell and single-channel electrophysiology of oocytes expressing Xenopus αβγ- or δβγ-ENaC, we demonstrate that the presence of the δ-subunit enhances the amount of current generated by ENaC due to an increased open probability, but also changes current into a transient form. Activity of canonical ENaCs is critically dependent on proteolytic processing of the α- and γ-subunits, and immunoblotting with epitope-tagged ENaC subunits indicated that, unlike α-ENaC, the δ-subunit does not undergo proteolytic maturation by the endogenous protease furin. Furthermore, currents generated by δβγ-ENaC were insensitive to activation by extracellular chymotrypsin, and presence of the δ-subunit prevented cleavage of γ-ENaC at the cell surface. Our findings suggest that subunit composition constitutes an additional level of ENaC regulation, and we propose that the Xenopus δ-ENaC subunit represents a functional example that demonstrates the importance of proteolytic maturation during ENaC evolution.
Collapse
Affiliation(s)
- Lukas Wichmann
- From the School of Natural and Environmental Sciences, Newcastle University, Ridley Building 2, Newcastle upon Tyne NE1 7RU, United Kingdom and.,the Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Kirsty Sophia Vowinkel
- the Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Alexander Perniss
- the Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Ivan Manzini
- the Institute of Animal Physiology, Department of Animal Physiology and Molecular Biomedicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Mike Althaus
- From the School of Natural and Environmental Sciences, Newcastle University, Ridley Building 2, Newcastle upon Tyne NE1 7RU, United Kingdom and
| |
Collapse
|
38
|
Localization of epithelial sodium channel (ENaC) and CFTR in the germinal epithelium of the testis, Sertoli cells, and spermatozoa. J Mol Histol 2018; 49:195-208. [PMID: 29453757 DOI: 10.1007/s10735-018-9759-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/05/2018] [Indexed: 12/23/2022]
Abstract
Spermatogenesis starts within the seminiferous tubules of the testis by mitotic division of spermatogonia that produces spermatocytes. Meiotic division of these spermatocytes produces haploid spermatids that differentiate into spermatozoa. In this study, we examined the expression of ENaC and CFTR (a Cl- channel) in rat testicular sections using confocal microscopic immunofluorescence. The structural integrity of the seminiferous tubule sections was verified by precise phalloidin staining of the actin fibers located abundantly at both basal and adluminal tight junctions. The acrosome forming regions in the round spermatids were stained using an FITC coupled lectin (wheat germ agglutinin). In all phases of the germ cells (spermatogonia, spermatocytes, and spermatids) ENaC was localized in cytoplasmic pools. Prior to spermiation, ENaC immunofluorescence appeared along the tails of the spermatids. In spermatozoa isolated from the epididymis, ENaC was localized at the acrosome and a central region of the sperm flagellum. The mature sperm are transcriptionally silent. Hence, we suggest that ENaC subunits in cytoplasmic pools in germ cells serve as the source of ENaC subunits located along the tail of spermatozoa. The locations of ENaC is compatible with a possible role in the acrosomal reaction and sperm mobility. In contrast to ENaC, CFTR immunofluorescence was most strongly observed specifically within the Sertoli cell nuclei. Based on the nuclear localization of CFTR we suggest that, in addition to its role as an ion channel, CFTR may have an independent role in gene regulation within the nuclei.
Collapse
|
39
|
Antinucci M, Risso D. A Matter of Taste: Lineage-Specific Loss of Function of Taste Receptor Genes in Vertebrates. Front Mol Biosci 2017; 4:81. [PMID: 29234667 PMCID: PMC5712339 DOI: 10.3389/fmolb.2017.00081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/13/2017] [Indexed: 12/02/2022] Open
Abstract
Vertebrates can perceive at least five different taste qualities, each of which is thought to have a specific role in the evolution of different species. The avoidance of potentially poisonous foods, which are generally bitter or sour tasting, and the search for more nutritious ones, those with high-fat and high-sugar content, are two of the most well-known examples. The study of taste genes encoding receptors that recognize ligands triggering taste sensations has helped to reconstruct several evolutionary adaptations to dietary changes. In addition, an increasing number of studies have focused on pseudogenes, genomic DNA sequences that have traditionally been considered defunct relatives of functional genes mostly because of the presence of deleterious mutations interrupting their open reading frames. The study of taste receptor pseudogenes has helped to shed light on how the evolutionary history of taste in vertebrates has been the result of a succession of gene gain and loss processes. This dynamic role in evolution has been explained by the "less-is-more" hypothesis, suggesting gene loss as a mechanism of evolutionary change in response to a dietary shift. This mini-review aims at depicting the major lineage-specific loss of function of taste receptor genes in vertebrates, stressing their evolutionary importance and recapitulating signatures of natural selection and their correlations with food habits.
Collapse
Affiliation(s)
| | - Davide Risso
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
40
|
Bartoszewski R, Matalon S, Collawn JF. Ion channels of the lung and their role in disease pathogenesis. Am J Physiol Lung Cell Mol Physiol 2017; 313:L859-L872. [PMID: 29025712 PMCID: PMC5792182 DOI: 10.1152/ajplung.00285.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
Maintenance of normal epithelial ion and water transport in the lungs includes providing a thin layer of surface liquid that coats the conducting airways. This airway surface liquid is critical for normal lung function in a number of ways but, perhaps most importantly, is required for normal mucociliary clearance and bacterial removal. Preservation of the appropriate level of hydration, pH, and viscosity for the airway surface liquid requires the proper regulation and function of a battery of different types of ion channels and transporters. Here we discuss how alterations in ion channel/transporter function often lead to lung pathologies.
Collapse
Affiliation(s)
- Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
- Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
- Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
- Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
41
|
Hanukoglu I, Boggula VR, Vaknine H, Sharma S, Kleyman T, Hanukoglu A. Expression of epithelial sodium channel (ENaC) and CFTR in the human epidermis and epidermal appendages. Histochem Cell Biol 2017; 147:733-748. [PMID: 28130590 DOI: 10.1007/s00418-016-1535-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
A major function of the skin is the regulation of body temperature by sweat secretions. Sweat glands secrete water and salt, especially NaCl. Excreted water evaporates, cooling the skin surface, and Na+ ions are reabsorbed by the epithelial sodium channels (ENaC). Mutations in ENaC subunit genes lead to a severe multi-system (systemic) form of pseudohypoaldosteronism (PHA) type I, characterized by salt loss from aldosterone target organs, including sweat glands in the skin. In this study, we mapped the sites of localization of ENaC in the human skin by confocal microscopy using polyclonal antibodies generated against human αENaC. Our results reveal that ENaC is expressed strongly in all epidermal layers except stratum corneum, and also in the sebaceous glands, eccrine glands, arrector pili smooth muscle cells, and intra-dermal adipocytes. In smooth muscle cells and adipocytes, ENaC is co-localized with F-actin. No expression of ENaC was detected in the dermis. CFTR is strongly expressed in sebaceous glands. In epidermal appendages noted, except the eccrine sweat glands, ENaC is mainly located in the cytoplasm. In the eccrine glands and ducts, ENaC and CFTR are located on the apical side of the membrane. This localization of ENaC is compatible with ENaC's role in salt reabsorption. PHA patients may develop folliculitis, miliaria rubra, and atopic dermatitis-like skin lesions, due to sweat gland duct occlusion and inflammation of eccrine glands as a result of salt accumulation.
Collapse
Affiliation(s)
- Israel Hanukoglu
- Laboratory of Cell Biology, Ariel University, Ariel, 40700, Israel.
| | - Vijay R Boggula
- Laboratory of Cell Biology, Ariel University, Ariel, 40700, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Hananya Vaknine
- Division of Pathology, E. Wolfson Medical Center, Holon, Israel
| | - Sachin Sharma
- Laboratory of Cell Biology, Ariel University, Ariel, 40700, Israel
| | - Thomas Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron Hanukoglu
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Division of Pediatric Endocrinology, E. Wolfson Medical Center, Holon, Israel
| |
Collapse
|
42
|
Rauh R, Hoerner C, Korbmacher C. δβγ-ENaC is inhibited by CFTR but stimulated by cAMP in Xenopus laevis oocytes. Am J Physiol Lung Cell Mol Physiol 2016; 312:L277-L287. [PMID: 27941075 DOI: 10.1152/ajplung.00375.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 11/22/2022] Open
Abstract
The epithelial sodium channel (ENaC) and the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel critically regulate airway surface liquid by driving fluid absorption and secretion, respectively. Their functional interplay is complex and incompletely understood. ENaC is a heteromeric channel with three well-characterized subunits (α, β, and γ). In humans, an additional δ-ENaC subunit exists in lung and several other tissues, where it may replace the α-subunit to form δβγ-ENaC. Little is known about the physiological role of δβγ-ENaC and its possible interaction with CFTR. The aim of the present study was to investigate the effect of human CFTR on human δβγ-ENaC heterologously expressed in Xenopus laevis oocytes. In oocytes coexpressing δβγ-ENaC and CFTR the ENaC-mediated amiloride-sensitive whole cell current (ΔIami) was reduced by ~50% compared with that measured in oocytes expressing δβγ-ENaC alone. Moreover, basal level of proteolytic ENaC activation was reduced in the presence of CFTR. The inhibitory effect of CFTR on δβγ-ENaC was due to a combination of decreased average open probability (Po) and reduced channel expression at the cell surface. Interestingly, in oocytes expressing δβγ-ENaC, increasing intracellular [cAMP] by IBMX and forskolin increased ΔIami by ~50%. This stimulatory effect was not observed for human and rat αβγ-ENaC and was independent of CFTR coexpression and coactivation. Experiments with a mutant channel (δβS520Cγ-ENaC) which can be converted to a channel with a Po of nearly 1 suggested that cAMP activates δβγ-ENaC by increasing Po In conclusion, our results demonstrate that δβγ-ENaC is inhibited by CFTR but activated by cAMP.
Collapse
Affiliation(s)
- Robert Rauh
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian Hoerner
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
43
|
Hanukoglu I. ASIC and ENaC type sodium channels: conformational states and the structures of the ion selectivity filters. FEBS J 2016; 284:525-545. [DOI: 10.1111/febs.13840] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/04/2016] [Accepted: 08/26/2016] [Indexed: 12/18/2022]
|
44
|
Hanukoglu I, Hanukoglu A. Epithelial sodium channel (ENaC) family: Phylogeny, structure-function, tissue distribution, and associated inherited diseases. Gene 2016; 579:95-132. [PMID: 26772908 PMCID: PMC4756657 DOI: 10.1016/j.gene.2015.12.061] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/20/2015] [Accepted: 12/22/2015] [Indexed: 01/24/2023]
Abstract
The epithelial sodium channel (ENaC) is composed of three homologous subunits and allows the flow of Na(+) ions across high resistance epithelia, maintaining body salt and water homeostasis. ENaC dependent reabsorption of Na(+) in the kidney tubules regulates extracellular fluid (ECF) volume and blood pressure by modulating osmolarity. In multi-ciliated cells, ENaC is located in cilia and plays an essential role in the regulation of epithelial surface liquid volume necessary for cilial transport of mucus and gametes in the respiratory and reproductive tracts respectively. The subunits that form ENaC (named as alpha, beta, gamma and delta, encoded by genes SCNN1A, SCNN1B, SCNN1G, and SCNN1D) are members of the ENaC/Degenerin superfamily. The earliest appearance of ENaC orthologs is in the genomes of the most ancient vertebrate taxon, Cyclostomata (jawless vertebrates) including lampreys, followed by earliest representatives of Gnathostomata (jawed vertebrates) including cartilaginous sharks. Among Euteleostomi (bony vertebrates), Actinopterygii (ray finned-fishes) branch has lost ENaC genes. Yet, most animals in the Sarcopterygii (lobe-finned fish) branch including Tetrapoda, amphibians and amniotes (lizards, crocodiles, birds, and mammals), have four ENaC paralogs. We compared the sequences of ENaC orthologs from 20 species and established criteria for the identification of ENaC orthologs and paralogs, and their distinction from other members of the ENaC/Degenerin superfamily, especially ASIC family. Differences between ENaCs and ASICs are summarized in view of their physiological functions and tissue distributions. Structural motifs that are conserved throughout vertebrate ENaCs are highlighted. We also present a comparative overview of the genotype-phenotype relationships in inherited diseases associated with ENaC mutations, including multisystem pseudohypoaldosteronism (PHA1B), Liddle syndrome, cystic fibrosis-like disease and essential hypertension.
Collapse
Affiliation(s)
- Israel Hanukoglu
- Laboratory of Cell Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel.
| | - Aaron Hanukoglu
- Division of Pediatric Endocrinology, E. Wolfson Medical Center, Holon, Israel; Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
45
|
Kumarhia D, He L, McCluskey LP. Inflammatory stimuli acutely modulate peripheral taste function. J Neurophysiol 2016; 115:2964-75. [PMID: 27009163 DOI: 10.1152/jn.01104.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/23/2016] [Indexed: 12/30/2022] Open
Abstract
Inflammation-mediated changes in taste perception can affect health outcomes in patients, but little is known about the underlying mechanisms. In the present work, we hypothesized that proinflammatory cytokines directly modulate Na(+) transport in taste buds. To test this, we measured acute changes in Na(+) flux in polarized fungiform taste buds loaded with a Na(+) indicator dye. IL-1β elicited an amiloride-sensitive increase in Na(+) transport in taste buds. In contrast, TNF-α dramatically and reversibly decreased Na(+) flux in polarized taste buds via amiloride-sensitive and amiloride-insensitive Na(+) transport systems. The speed and partial amiloride sensitivity of these changes in Na(+) flux indicate that IL-1β and TNF-α modulate epithelial Na(+) channel (ENaC) function. A portion of the TNF-mediated decrease in Na(+) flux is also blocked by the TRPV1 antagonist capsazepine, although TNF-α further reduced Na(+) transport independently of both amiloride and capsazepine. We also assessed taste function in vivo in a model of infection and inflammation that elevates these and additional cytokines. In rats administered systemic lipopolysaccharide (LPS), CT responses to Na(+) were significantly elevated between 1 and 2 h after LPS treatment. Low, normally preferred concentrations of NaCl and sodium acetate elicited high response magnitudes. Consistent with this outcome, codelivery of IL-1β and TNF-α enhanced Na(+) flux in polarized taste buds. These results demonstrate that inflammation elicits swift changes in Na(+) taste function, which may limit salt consumption during illness.
Collapse
Affiliation(s)
- Devaki Kumarhia
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and Graduate Program in Molecular Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Lianying He
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| | - Lynnette Phillips McCluskey
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| |
Collapse
|
46
|
Plasma membrane insertion of epithelial sodium channels occurs with dual kinetics. Pflugers Arch 2016; 468:859-70. [DOI: 10.1007/s00424-016-1799-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 01/05/2023]
|
47
|
Matalon S, Bartoszewski R, Collawn JF. Role of epithelial sodium channels in the regulation of lung fluid homeostasis. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1229-38. [PMID: 26432872 DOI: 10.1152/ajplung.00319.2015] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/25/2015] [Indexed: 01/11/2023] Open
Abstract
In utero, fetal lung epithelial cells actively secrete Cl(-) ions into the lung air spaces while Na(+) ions follow passively to maintain electroneutrality. This process, driven by an electrochemical gradient generated by the Na(+)-K(+)-ATPase, is responsible for the secretion of fetal fluid that is essential for normal lung development. Shortly before birth, a significant upregulation of amiloride-sensitive epithelial channels (ENaCs) on the apical side of the lung epithelial cells results in upregulation of active Na(+) transport. This process is critical for the reabsorption of fetal lung fluid and the establishment of optimum gas exchange. In the adult lung, active Na(+) reabsorption across distal lung epithelial cells limits the degree of alveolar edema in patients with acute lung injury and cardiogenic edema. Cl(-) ions are transported either paracellularly or transcellularly to preserve electroneutrality. An increase in Cl(-) secretion across the distal lung epithelium has been reported following an acute increase in left atrial pressure and may result in pulmonary edema. In contrast, airway epithelial cells secrete Cl(-) through apical cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels and absorb Na(+). Thus the coordinated action of Cl(-) secretion and Na(+) absorption is essential for maintenance of the volume of epithelial lining fluid that, in turn, maximizes mucociliary clearance and facilitates clearance of bacteria and debris from the lungs. Any factor that interferes with Na(+) or Cl(-) transport or dramatically upregulates ENaC activity in airway epithelial cells has been associated with lung diseases such as cystic fibrosis or chronic obstructive lung disease. In this review we focus on the role of the ENaC, the mechanisms involved in ENaC regulation, and how ENaC dysregulation can lead to lung pathology.
Collapse
Affiliation(s)
- Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
48
|
Perniss A, Wolf A, Wichmann L, Schönberger M, Althaus M. Evans Blue is not a suitable inhibitor of the epithelial sodium channel δ-subunit. Biochem Biophys Res Commun 2015; 466:468-74. [PMID: 26365349 DOI: 10.1016/j.bbrc.2015.09.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 01/06/2023]
Abstract
The Epithelial Sodium Channel (ENaC) is a heterotrimeric ion channel which can be either formed by assembly of its α-, β- and γ-subunits or, alternatively, its δ-, β- and γ-subunits. The physiological function of αβγ-ENaC is well established, but the function of δβγ-ENaC remains elusive. The azo-dye Evans Blue (EvB) has been routinely used to discriminate between the two channel isoforms by decreasing transmembrane currents and amiloride-sensitive current fractions of δβγ-ENaC expressing Xenopus oocytes. Even though these results could be reproduced, it was found by precipitation experiments and spectroscopic methods that the cationic amiloride and the anionic EvB directly interact in solution, forming a strong complex. Thereby a large amount of pharmacologically available amiloride is removed from physiological buffer solutions and the effective amiloride concentration is reduced. This interaction did not occur in the presence of albumin. In microelectrode recordings, EvB was able to abrogate the block of δβγ-ENaC by amiloride or its derivative benzamil. In sum, EvB reduces amiloride-sensitive ion current fractions in electrophysiological experiments. This is not a result of a specific inhibition of δβγ-ENaC but rather represents a pharmacological artefact. EvB should therefore not be used as an inhibitor of δ-ENaC.
Collapse
Affiliation(s)
- Alexander Perniss
- Institute for Animal Physiology, Justus-Liebig-University, Heinrich-Buff-Ring 26, D-35392 Giessen, Germany
| | - Annemarie Wolf
- Institute for Animal Physiology, Justus-Liebig-University, Heinrich-Buff-Ring 26, D-35392 Giessen, Germany
| | - Lukas Wichmann
- Institute for Animal Physiology, Justus-Liebig-University, Heinrich-Buff-Ring 26, D-35392 Giessen, Germany
| | - Matthias Schönberger
- Department of Chemistry, Stanford University, 337 Campus Drive, Stanford, CA 94305-4401337, USA; Institute for Radiochemistry and Molecular Imaging, University Clinic Cologne, Kerpener Strasse 62, 50931 Cologne, Germany.
| | - Mike Althaus
- Institute for Animal Physiology, Justus-Liebig-University, Heinrich-Buff-Ring 26, D-35392 Giessen, Germany.
| |
Collapse
|
49
|
Elevated Expression of the Delta-Subunit of Epithelial Sodium Channel in Temporal Lobe Epilepsy Patients and Rat Model. J Mol Neurosci 2015; 57:510-8. [DOI: 10.1007/s12031-015-0630-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/23/2015] [Indexed: 01/13/2023]
|
50
|
Jeggle P, Smith ESJ, Stewart AP, Haerteis S, Korbmacher C, Edwardson JM. Atomic force microscopy imaging reveals the formation of ASIC/ENaC cross-clade ion channels. Biochem Biophys Res Commun 2015; 464:38-44. [PMID: 26032502 DOI: 10.1016/j.bbrc.2015.05.091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/28/2015] [Indexed: 01/02/2023]
Abstract
ASIC and ENaC are co-expressed in various cell types, and there is evidence for a close association between them. Here, we used atomic force microscopy (AFM) to determine whether ASIC1a and ENaC subunits are able to form cross-clade hybrid ion channels. ASIC1a and ENaC could be co-isolated from detergent extracts of tsA 201 cells co-expressing the two subunits. Isolated proteins were incubated with antibodies against ENaC and Fab fragments against ASIC1a. AFM imaging revealed proteins that were decorated by both an antibody and a Fab fragment with an angle of ∼120° between them, indicating the formation of ASIC1a/ENaC heterotrimers.
Collapse
Affiliation(s)
- Pia Jeggle
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Andrew P Stewart
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Silke Haerteis
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstrasse 6, 91054 Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Waldstrasse 6, 91054 Erlangen, Germany
| | - J Michael Edwardson
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom.
| |
Collapse
|