1
|
Gideon S, Boyd B, Ramirez Sierra B, Arenas D, Ochoa P, Eme J, Kolosov D. Voltage-gated ion channels in cultured gill epithelia of rainbow trout, Oncorhynchus mykiss, change in transcript abundance with exposure to freshwater. Comp Biochem Physiol A Mol Integr Physiol 2025; 304:111835. [PMID: 40074167 DOI: 10.1016/j.cbpa.2025.111835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Salmonid fishes are well adapted to transition between salinities as part of a diadromid lifestyle, and many species are both economically and environmentally important. Ion-transporting gill epithelium helps fishes maintain ion balance during salinity transition. Recent transcriptomic surveys suggest that voltage-gated ion channels (VGICs) are present in gill epithelium of fishes. However, fish gill epithelia are architecturally complex and structurally heterogeneous (which includes layers of excitable tissues), which necessitates a model to study isolated gill epithelial cells. In the present study, we isolated gill epithelial cells, used them to reconstruct primary cultured gill epithelium model, and exposed the reconstructed epithelia to apical freshwater (FW). Using RNAseq and molecular biology we demonstrate that multiple VGICs are expressed in cultured gill epithelia of a salmonid, rainbow trout Oncorhynchus mykiss. Following apical exposure to FW, multiple subunits of voltage-gated calcium (CaV) channels, as well as KCNE2 were upregulated in mRNA abundance. Using a custom-made antibody, we demonstrated that CaV1.3 immunolocalized to the apical membrane of epithelia in intact trout gill, as well as in the cultured gill epithelium. Pharmacological inhibition of CaV1 in FW-exposed cultured epithelia led to increased transepithelial resistance. Therefore, we propose that VGICs are present in gill epithelia of fishes, and may rapidly and autonomously respond to environmental salinity changes to help the fish maintain salt and water balance, where CaV1 specifically may play a particularly important role in rapid adjustment of gill epithelia barrier properties and resistivity and potentially in responding to regulatory cell volume decrease in vitro.
Collapse
Affiliation(s)
- Siaje Gideon
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Brendan Boyd
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Brandon Ramirez Sierra
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Dennise Arenas
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Perla Ochoa
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - John Eme
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA
| | - Dennis Kolosov
- Department of Biological Sciences, California State University San Marcos, CA 92096, USA.
| |
Collapse
|
2
|
Englisch CN, Diebolt CM, Kirstein E, Wahl V, Wartenberg P, Schaudien D, Beckmann A, Laschke MW, Krasteva-Christ G, Gudermann T, Chubanov V, Boehm U, Tschernig T. TRPM6 in murine kidneys-of targets and antibodies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03951-0. [PMID: 40025338 DOI: 10.1007/s00210-025-03951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Magnesium is the fourth most abundant cation in the human organism. As a key-player in many enzymatic reactions, magnesium homeostasis disbalance can cause severe disorders. In the early 2000s, the transient receptor potential melastatin channel 6 (TRPM6) was identified as a critical protein in renal Mg2+-reabsorption in the distal convoluted tubule (DCT). As the key-interface responsible for salt/water adaptation to environmental changes, the kidney is a highly dynamic system. Therefore, renal TRPM6 expression and Mg2+-reabsorption might not be restricted to the DCT, as previously indicated. To address this, protein targeting is mandatory since genomic detection is insufficient to conclude on functional expression. For this purpose, we used a polyclonal TRPM6 antibody from an established manufacturer and detected immunostaining in murine proximal and distal tubules. As a matter of fact, the specificity of most commercially available TRPM6 antibodies is insufficiently validated which relies on the lack of constitutive trpm6 knockouts. Therefore, conditional trpm6 knockout mice were used for control experiments. Similar signals were observed in the knockout tissue when compared to wildtype using the TRPM6 antibody. Overlaps with TRPM7 epitopes or other peptides are conceivable. Thus, TRPM6 immunohistochemistry and immunofluorescence results are difficult to interpret, and the spectrum of renal TRPM6 expression is not yet elucidated.
Collapse
Affiliation(s)
- Colya N Englisch
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany
| | - Coline M Diebolt
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany
| | - Emilie Kirstein
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany
| | - Vanessa Wahl
- Institute of Pharmacology, Saarland University, 66421, Homburg/Saar, Germany
| | - Philipp Wartenberg
- Institute of Pharmacology, Saarland University, 66421, Homburg/Saar, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625, Hannover, Germany
| | - Anja Beckmann
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
| | - Ulrich Boehm
- Institute of Pharmacology, Saarland University, 66421, Homburg/Saar, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Strasse 100, 66421, Homburg/Saar, Germany.
| |
Collapse
|
3
|
Kocabey Sütçü Z, Aytaç Kaplan EH, Önal H. Pediatric hypoparathyroidism: etiological and clinical evaluation in a tertiary center. Endocrine 2025; 87:1235-1245. [PMID: 39586905 DOI: 10.1007/s12020-024-04110-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE This study aims to evaluate the etiology, clinical presentation, and management of pediatric hypoparathyroidism in a tertiary center. METHODS A retrospective review was conducted on pediatric patients diagnosed with hypoparathyroidism at the Pediatric Endocrinology Clinic from March 2021 to June 2023. Data on demographic characteristics, presenting symptoms, laboratory findings, genetic analyses, and treatment outcomes were collected. RESULTS A total of 56 patients (31 females) were included. The median age at diagnosis of the patients was 5.5 years (range 0.04-17 years), and the median age of symptom onset was 5 years (range 0.04-16.5 years). The etiology was genetic and idiopathic in 39 patients (70.9%), with syndromic forms, familial isolated hypoparathyroidism, and hypomagnesemia identified. DiGeorge syndrome was present in 14 patients, making it the most common syndromic form. The syndromes associated with hypomagnesemia were those with mutations in the TRMP6 and CLDN16 genes. Sixteen patients (29.1%) had acquired causes, primarily post-thyroid surgery and autoimmune conditions. Common symptoms included muscle spasms (32.7%) and seizures (21.8%). Laboratory findings revealed a median serum calcium level of 6.7 mg/dL (3.8-8.5) and median serum phosphorus level of 7.7 (4.9-12.5) mg/dL. Treatment primarily involved calcitriol [The median dose of calcitriol is 25 ng/kg/day (range: 25-50 ng/kg/day)] and calcium [The median dose of calcium gluconate is 0.7 mL/kg (range: 0.5-1 mL/kg) and oral calcium is 1000 mg (range: 700-1300 mg)] supplementation. Intravenous calcium gluconate treatment was administered to 39 (70.6%) patients, oral calcium carbonate therapy was given to 16 (29.1%) patients, and calcitriol treatment was initiated for 51 (91.1%) patients. Complications such as nephrocalcinosis (7, 13.7%) and hypercalciuria (7, 13.7%) were observed in some patients. CONCLUSION This study emphasizes the significant genetic component, particularly syndromic, in pediatric hypoparathyroidism, highlighting the need for comprehensive genetic evaluation and a multidisciplinary approach for effective management, especially concerning complications. In this way, early and accurate diagnosis will reduce unnecessary tests, treatment approaches, and repeated hospital visits. Regular monitoring is essential to mitigate potential complications associated with long-term treatment.
Collapse
Affiliation(s)
- Zümrüt Kocabey Sütçü
- Department of Pediatric Endocrinology, Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye.
| | - Emel Hatun Aytaç Kaplan
- Department of Pediatric Endocrinology, Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye
| | - Hasan Önal
- Department of Pediatric Endocrinology, Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye
| |
Collapse
|
4
|
Stumpff F, Manneck D. Prebiotics as modulators of colonic calcium and magnesium uptake. Acta Physiol (Oxf) 2025; 241:e14262. [PMID: 39803707 PMCID: PMC11726438 DOI: 10.1111/apha.14262] [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: 07/18/2024] [Revised: 11/23/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Ca2+ and Mg2+ are essential nutrients, and deficiency can cause serious health problems. Thus, lack of Ca2+ and Mg2+ can lead to osteoporosis, with incidence rising both in absolute and age-specific terms, while Mg2+ deficiency is associated with type II diabetes. Prevention via vitamin D or estrogen is controversial, and the bioavailability of Ca2+ and Mg2+ from supplements is significantly lower than that from milk products. Problems are likely to increase as populations age and the number of people on vegan diets surges. Developing new therapeutic strategies requires a better understanding of the molecular mechanisms involved in absorption by intestinal epithelia. The vitamin-D dependent, active pathway for the uptake of Ca2+ from the upper small intestine involving TRPV6 is highly efficient but only accounts for about 20% of total uptake. Instead, most Ca2+ uptake is thought to occur via passive paracellular diffusion across the ileum, although sufficiently high luminal concentrations are difficult to achieve.. Interestingly, colon and caecum also have a considerable capacity for the active absorption of Ca2+ and Mg2+, the molecular mechanisms of which are unclear. Intriguingly, stimulating fermentation by prebiotics enhances colonic absorption, which can rise from ~10% to ~30% of the total. Notably, fermentation releases protons, which inhibits channels highly selective for Ca2+ and Mg2+ (TRPV6 and TRPM6/TRPM7). Conversely, the non-selective cation channel TRPV3 is stimulated by both intracellular acidification and by numerous herbal compounds. Spicy, fiber-rich food, as traditionally consumed in many cultures, might enhance the uptake of Ca2+ and Mg2+ via this pathway.
Collapse
Affiliation(s)
- Friederike Stumpff
- Institute for Molecular MedicineHealth and Medical University PotsdamPotsdamGermany
| | - David Manneck
- Institute for Molecular MedicineHealth and Medical University PotsdamPotsdamGermany
| |
Collapse
|
5
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025; 125:660-744. [PMID: 39746035 PMCID: PMC11758815 DOI: 10.1021/acs.chemrev.4c00577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/10/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université
Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
6
|
Adella A, Gommers LMM, Bos C, Leermakers PA, de Baaij JHF, Hoenderop JGJ. Characterization of intestine-specific TRPM6 knockout C57BL/6 J mice: effects of short-term omeprazole treatment. Pflugers Arch 2025; 477:99-109. [PMID: 39266724 PMCID: PMC11711252 DOI: 10.1007/s00424-024-03017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
The transient receptor potential melastatin type 6 (TRPM6) is a divalent cation channel pivotal for gatekeeping Mg2+ balance. Disturbance in Mg2+ balance has been associated with the chronic use of proton pump inhibitors (PPIs) such as omeprazole. In this study, we investigated if TRPM6 plays a role in mediating the effects of short-term (4 days) omeprazole treatment on intestinal Mg2+ malabsorption using intestine-specific TRPM6 knockout (Vill1-TRPM6-/-) mice. To do this, forty-eight adult male C57BL/6 J mice (50% TRPM6fl/fl and 50% Vill1-TRPM6-/-) were characterized, and the distal colon of these mice was subjected to RNA sequencing. Moreover, these mice were exposed to 20 mg/kg bodyweight omeprazole or placebo for 4 days. Vill1-TRPM6-/- mice had a significantly lower 25Mg2+ absorption compared to control TRPM6fl/fl mice, accompanied by lower Mg2+ serum levels, and urinary Mg2+ excretion. Furthermore, renal Slc41a3, Trpm6, and Trpm7 gene expressions were higher in these animals, indicating a compensatory mechanism via the kidney. RNA sequencing of the distal colon revealed a downregulation of the Mn2+ transporter Slc30a10. However, no changes in Mn2+ serum, urine, and feces levels were observed. Moreover, 4 days omeprazole treatment did not affect Mg2+ homeostasis as no changes in serum 25Mg2+ and total Mg2+ were seen. In conclusion, we demonstrate here for the first time that Vill1-TRPM6-/- mice have a lower Mg2+ absorption in the intestines. Moreover, short-term omeprazole treatment does not alter Mg2+ absorption in both Vill1-TRPM6-/- and TRPM6fl/fl mice. This suggests that TRPM6-mediated Mg2+ absorption in the intestines is not affected by short-term PPI administration.
Collapse
Affiliation(s)
- Anastasia Adella
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisanne M M Gommers
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter A Leermakers
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
7
|
Brodnanova M, Cibulka M, Grendar M, Gondas E, Kolisek M. IL-6 Does Not Influence the Expression of SLC41A1 and Other Mg-Homeostatic Factors. Int J Mol Sci 2024; 25:13274. [PMID: 39769039 PMCID: PMC11675721 DOI: 10.3390/ijms252413274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Together with chronic inflammation, disturbed magnesium homeostasis is a factor accompanying chronic disease which thus contributes to a reduced quality of human life. In this study, our objective was to examine the possible IL-6-mediated chronic inflammation-dependent regulation of nine magnesiotropic genes encoding for constituents of magnesium homeostasis of the cell. We used three cell lines (HepG2, U-266, and PANC-1), all characterized by high expression of the IL6R gene and the presence of a membrane form of IL-6R capable of responding to human IL-6. Despite the confirmed activation of the IL-6R/JAK/STAT3 pathway after hIL-6 treatment, we observed no biologically relevant changes in the transcription intensity of the studied magnesiotropic genes. This, however, does not exclude the possibility that IL-6 can affect magnesium homeostasis at levels other than through modified transcription.
Collapse
Affiliation(s)
- Maria Brodnanova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 03601 Martin, Slovakia; (M.B.); (M.C.); (M.G.)
| | - Michal Cibulka
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 03601 Martin, Slovakia; (M.B.); (M.C.); (M.G.)
| | - Marian Grendar
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 03601 Martin, Slovakia; (M.B.); (M.C.); (M.G.)
| | - Eduard Gondas
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 03601 Martin, Slovakia;
| | - Martin Kolisek
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, 03601 Martin, Slovakia; (M.B.); (M.C.); (M.G.)
| |
Collapse
|
8
|
Takvam M, Denker E, Gharbi N, Tronci V, Kolarevic J, Nilsen TO. Differential regulation of magnesium transporters Slc41, Cnnm and Trpm6-7 in the kidney of salmonids may represent evolutionary adaptations to high salinity environments. BMC Genomics 2024; 25:1156. [PMID: 39614204 PMCID: PMC11605958 DOI: 10.1186/s12864-024-11055-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
Magnesium is important for enzymatic reactions and physiological functions, and its intracellular concentration is tightly regulated. Atlantic salmon has the ability to handle large changes in environmental Mg2+ concentration when migrating between freshwater and seawater habitats, making it a relevant model to investigate Mg2+ homeostasis. Parr-smolt transformation (PST) is a life history transition which prepares the freshwater juvenile for the marine environment. The kidney is one of the key organs involved in handling higher salt load in teleosts. Though several key Mg2+ transport families (SLC41, CNNM and TRPM6-7) have recently been identified in mammals and a few fishes, the molecular bases of Mg2+ homeostasis in salmon are not known. We found that all three families are represented in the salmon genome and exhibit a clear conservation of key functional domains and residues. Present study indicates a selective retention of paralogous Mg2+ transporters from the fourth whole genome duplication round (Ss4R) and a differential regulation of these genes, which suggests neo- and/or sub-functionalization events. slc41a1-1, cnnm4a1, -4a2 and trpm7-2 are the main upregulated genes in the kidney during PST and remain high or further increase after exposure to seawater (33 ppt). By contrast, slc41a1-2, -3a, cnnm3-1, and cnnm3-2 are only upregulated after seawater exposure. In addition, slc41a1-1, -2, and trpm7-2 respond when exposed to brackish water (12 ppt), while cnnm3-1 and cnnm3-2 do not, indicating the existence of a lower salinity threshold response for these members. Finally, the response of slc41a1-1, -2 and trpm7-2 in salmon was significantly reduced or completely abolished when exposed to Mg2+-reduced brackish water, while others were not, suggesting they might be specifically regulated by Mg2+. Our results are consistent with previous findings on other euryhaline teleosts and chondrichthyan species, suggesting the existence of common adaptive strategies to thrive in high salinity environments. Concomitantly, salmonid-specific innovations, such as differential regulation and recruitment of family members not previously shown to be regulated in the kidney (Cnnm1 and Cnnm4) of other vertebrates might point to adaptions associated with their very plastic anadromous life cycle.
Collapse
Affiliation(s)
- Marius Takvam
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
- NORCE, Norwegian Research Center, NORCE Environment and Climate, Bergen, Norway.
| | - Elsa Denker
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Naouel Gharbi
- NORCE, Norwegian Research Center, NORCE Environment and Climate, Bergen, Norway
| | - Valentina Tronci
- NORCE, Norwegian Research Center, NORCE Environment and Climate, Bergen, Norway
| | - Jelena Kolarevic
- Faculty of Biosciences, Fisheries and Economics, The Arctic University of Norway, Tromsø, 9037, Norway
| | - Tom Ole Nilsen
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| |
Collapse
|
9
|
Carnegie RE, Zheng J, Borges MC, Jones HJ, Wade KH, Sallis HM, Lewis SJ, Evans DM, Revez JA, The Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium, Evans J, Martin RM. Micronutrients and Major Depression: A Mendelian Randomisation Study. Nutrients 2024; 16:3690. [PMID: 39519523 PMCID: PMC11547740 DOI: 10.3390/nu16213690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Various vitamins and minerals have been implicated in the aetiology of depression. OBJECTIVE To estimate the effects of micronutrient exposures on major depressive disorder (MDD) and recurrent depression (rMDD) using Mendelian randomisation (MR), a method using genetic data to estimate causal effects given certain assumptions. METHODS We undertook a comprehensive bidirectional MR study of multiple micronutrient exposures on MDD and rMDD. Summary statistics were obtained from the Psychiatric Genomics Consortium (PGC) genome-wide association studies (GWASs) of MDD (cases = 116,209; controls = 314,566) and rMDD (cases = 17,451; controls = 62,482). RESULTS None of the micronutrients with available genetic instruments were strongly associated with MDD or rMDD using traditional MR methods. However, using methods to increase analytical power by accounting for genetically correlated variants (e.g., cIVW) highlighted five micronutrients with possible causal effects. Point estimates for rMDD were the largest magnitude, with three micronutrients suggestive of a protective effect: serum iron (ORcIVW 0.90 per SD increase; 95% CI 0.85-0.95; p = 0.0003); erythrocyte copper (ORcIVW 0.97; 95% CI 0.95-0.99; p = 0.0004); and 25(OH) vitamin D (ORcIVW 0.81; 0.66-0.99; p = 0.04). Apparent adverse effects of increased selenium on the risk of MDD (ORcIVW 1.03; 95% CI 1.02-1.05; p = 0.0003) and rMDD (ORcIVW 1.08; 95% CI 1.00-1.08; p = 0.06), and serum magnesium on rMDD (ORcIVW 1.21; 1.01-1.44; p = 0.04); were less consistent between methods and may be driven by pleiotropy. CONCLUSIONS Our results suggest weak evidence for a protective effect of iron, copper and 25(OH)D on major depressive outcomes, with mixed evidence for selenium and magnesium. There was no evidence to support a causal effect of any other micronutrients on MDD or rMDD, although genetic instruments were lacking, with insufficient power to detect small but important effects. Future micronutrient supplementation trials should ensure ample statistical power given modest causal effect estimates and consider potential risks of supplementation, as some micronutrient effect estimates suggested potential harm in excess.
Collapse
Affiliation(s)
- Rebecca E. Carnegie
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK (J.E.)
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Jie Zheng
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Maria C. Borges
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Hannah J. Jones
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK (J.E.)
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- NIHR Biomedical Research Centre, a Partnership between University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Kaitlin H. Wade
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Hannah M. Sallis
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK (J.E.)
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
- School of Psychological Science, University of Bristol, Bristol BS8 1TU, UK
| | - Sarah J. Lewis
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - David M. Evans
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD 4102, Australia
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joana A. Revez
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Jonathan Evans
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK (J.E.)
| | - Richard M. Martin
- Medical Research Centre (MRC), Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
- NIHR Biomedical Research Centre, a Partnership between University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| |
Collapse
|
10
|
Bosman W, Butler KM, Chang CA, Ganapathi M, Guzman E, Latta F, Chung WK, Claverie-Martin F, Davis JM, Hoenderop JGJ, de Baaij JHF. Pathogenic heterozygous TRPM7 variants and hypomagnesemia with developmental delay. Clin Kidney J 2024; 17:sfae211. [PMID: 39099563 PMCID: PMC11295107 DOI: 10.1093/ckj/sfae211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Indexed: 08/06/2024] Open
Abstract
Background Heterozygous variants in Transient receptor potential melastatin type 7 (TRPM7), encoding an essential and ubiquitously expressed cation channel, may cause hypomagnesemia, but current evidence is insufficient to draw definite conclusions and it is unclear whether any other phenotypes can occur. Methods Individuals with unexplained hypomagnesemia underwent whole-exome sequencing which identified TRPM7 variants. Pathogenicity of the identified variants was assessed by combining phenotypic, functional and in silico analyses. Results We report three new heterozygous missense variants in TRPM7 (p.Met1000Thr, p.Gly1046Arg, p.Leu1081Arg) in individuals with hypomagnesemia. Strikingly, autism spectrum disorder and developmental delay, mainly affecting speech and motor skills, was observed in all three individuals, while two out of three also presented with seizures. The three variants are predicted to be severely damaging by in silico prediction tools and structural modeling. Furthermore, these variants result in a clear loss-of-function of TRPM7-mediated magnesium uptake in vitro, while not affecting TRPM7 expression or insertion into the plasma membrane. Conclusions This study provides additional evidence for the association between heterozygous TRPM7 variants and hypomagnesemia and adds developmental delay to the phenotypic spectrum of TRPM7-related disorders. Considering that the TRPM7 gene is relatively tolerant to loss-of-function variants, future research should aim to unravel by what mechanisms specific heterozygous TRPM7 variants can cause disease.
Collapse
Affiliation(s)
- Willem Bosman
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
| | | | - Caitlin A Chang
- Department of Medical Genetics, BC Women and Children's Hospital, Vancouver, British Columbia, Canada
| | - Mythily Ganapathi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Edwin Guzman
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Femke Latta
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Felix Claverie-Martin
- Unidad de Investigación, RenalTube Group, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | | | | |
Collapse
|
11
|
Staruschenko A, Alexander RT, Caplan MJ, Ilatovskaya DV. Calcium signalling and transport in the kidney. Nat Rev Nephrol 2024; 20:541-555. [PMID: 38641658 PMCID: PMC12036682 DOI: 10.1038/s41581-024-00835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 04/21/2024]
Abstract
The kidney plays a pivotal role in regulating calcium levels within the body. Approximately 98% of the filtered calcium is reabsorbed in the nephron, and this process is tightly controlled to maintain calcium homeostasis, which is required to facilitate optimal bone mineralization, preserve serum calcium levels within a narrow range, and support intracellular signalling mechanisms. The maintenance of these functions is attributed to a delicate balance achieved by various calcium channels, transporters, and calcium-binding proteins in renal cells. Perturbation of this balance due to deficiency or dysfunction of calcium channels and calcium-binding proteins can lead to severe complications. For example, polycystic kidney disease is linked to aberrant calcium transport and signalling. Furthermore, dysregulation of calcium levels can promote the formation of kidney stones. This Review provides an updated description of the key aspects of calcium handling in the kidney, focusing on the function of various calcium channels and the physiological stimuli that control these channels or are communicated through them. A discussion of the role of calcium as an intracellular second messenger and the pathophysiology of renal calcium dysregulation, as well as a summary of gaps in knowledge and future prospects, are also included.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA.
- James A. Haley Veterans Hospital, Tampa, FL, USA.
| | - R Todd Alexander
- Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
- Women's and Children's Health Institute, Edmonton, AB, Canada
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
12
|
Touyz RM, de Baaij JHF, Hoenderop JGJ. Magnesium Disorders. N Engl J Med 2024; 390:1998-2009. [PMID: 38838313 DOI: 10.1056/nejmra1510603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Affiliation(s)
- Rhian M Touyz
- From the Research Institute of McGill University Health Centre, Departments of Medicine and Family Medicine, McGill University, Montreal (R.M.T.); and the Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands (J.H.F.B., J.G.J.H.)
| | - Jeroen H F de Baaij
- From the Research Institute of McGill University Health Centre, Departments of Medicine and Family Medicine, McGill University, Montreal (R.M.T.); and the Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands (J.H.F.B., J.G.J.H.)
| | - Joost G J Hoenderop
- From the Research Institute of McGill University Health Centre, Departments of Medicine and Family Medicine, McGill University, Montreal (R.M.T.); and the Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands (J.H.F.B., J.G.J.H.)
| |
Collapse
|
13
|
Xu L, Yuan P, Liu W, Liu L, Li X, Xie L. Magnesium status modulating the effect of serum vitamin D levels on retinopathy: National Health and Nutrition Examination Survey 2005 to 2008. Front Nutr 2024; 11:1408497. [PMID: 38895658 PMCID: PMC11183295 DOI: 10.3389/fnut.2024.1408497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Aim Magnesium levels may influence the effect of vitamin D levels on the body. This study aimed to assess the combined effect of magnesium status as reflected by magnesium depletion score (MDS) and vitamin D status on the risk of retinopathy. Methods This cross-sectional study included participants aged 40 years and older with complete information on vitamin D, MDS, and retinopathy assessment from the 2005-2008 National Health and Nutrition Examination Survey (NHANES). Logistic regression analysis was utilized to analyze the relationship of MDS and vitamin D with retinopathy and expressed as odds ratio (OR) and 95% confidence interval (CI). Results Of these 4,953 participants included, 602 (9.53%) participants had retinopathy. Serum vitamin D levels ≤30 nmol/L (vs. >30 nmol/L) (OR = 1.38, 95%CI: 1.05-1.81) and MDS >2 points (vs. ≤2 points) (OR = 1.47, 95%CI: 1.01-2.16) were associated with higher odds of retinopathy. There was an interaction between MDS and vitamin D on the increased odds of retinopathy (OR = 2.29, 95%CI: 1.12-4.68, P interaction = 0.025). In different MDS groups, serum vitamin D levels ≤30 nmol/L increased the odds of retinopathy only in the MDS >2 group (OR = 2.90, 95%CI: 1.16-7.24), but not in the MDS ≤2 group (p = 0.293). Subgroups analyses demonstrated that the interaction between MDS and serum vitamin D on retinopathy was observed in males (OR = 6.88, 95%CI: 1.41-33.66, P interaction = 0.019), people with diabetes (OR = 3.43, 95%CI: 1.78-6.63, P interaction < 0.001), and people with body mass index (BMI) ≥25 kg/m2 (OR = 2.46, 95%CI: 1.11-5.44, P interaction = 0.028). Conclusion Magnesium plays a moderating role in the relationship between serum vitamin D and retinopathy. The protective effect of vitamin D against retinopathy was primarily present among those with inadequate magnesium levels.
Collapse
Affiliation(s)
- Lei Xu
- Department of Ophthalmology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Penghua Yuan
- Department of Ophthalmology, Yudu County People’s Hospital, Ganzhou, Jiangxi, China
| | - Wanrong Liu
- Department of Ophthalmology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Linlin Liu
- Department of Ophthalmology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiongfeng Li
- Department of Ophthalmology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lianfeng Xie
- Department of Ophthalmology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
14
|
Shah CV, Sparks MA, Lee CT. Sodium/Glucose Cotransporter 2 Inhibitors and Magnesium Homeostasis: A Review. Am J Kidney Dis 2024; 83:648-658. [PMID: 38372686 DOI: 10.1053/j.ajkd.2023.11.006] [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: 08/08/2023] [Revised: 10/21/2023] [Accepted: 11/11/2023] [Indexed: 02/20/2024]
Abstract
Magnesium (Mg2+), also known as "the forgotten ion," is the second most abundant intracellular cation and is essential in a broad range of intracellular physiological and biochemical reactions. Its deficiency, hypomagnesemia (Mg2+<1.8mg/dL), is a prevalent condition and routinely poses challenges in its management in clinical practice. Sodium/glucose cotransporter 2 (SGLT2) inhibitors have emerged as a new class of drugs with treating hypomagnesemia as their unique extraglycemic benefit. The beneficial effect of SGLT2 inhibitors on magnesium balance in patients with diabetes with or without hypomagnesemia has been noted as a class effect in recent meta-analysis data from randomized clinical trials. Some reports have demonstrated their role in treating refractory hypomagnesemia in patients with or without diabetes. Moreover, studies on animal models have attempted to illustrate the effect of SGLT2 inhibitors on Mg2+homeostasis. In this review, we discuss the current evidence and possible pathophysiological mechanisms, and we provide directions for further research. We conclude by suggesting the effect of SGLT2 inhibitors on Mg2+homeostasis is a class effect, with certain patients gaining significant benefits. Further studies are needed to examine whether SGLT2 inhibitors can become a desperately needed novel class of medicines in treating hypomagnesemia.
Collapse
Affiliation(s)
- Chintan V Shah
- Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida.
| | - Matthew A Sparks
- Division of Nephrology and Department of Medicine, Duke University, and Durham VA Health Care System, Durham, North Carolina
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Kaohsiung Municipal Feng-Shan Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
15
|
Nie M, Zhang J, Bal M, Duran C, An SW, Zigman JM, Baum M, Hiremath C, Marciano DK, Wolf MTF. Ghrelin enhances tubular magnesium absorption in the kidney. Front Physiol 2024; 15:1363708. [PMID: 38638279 PMCID: PMC11024433 DOI: 10.3389/fphys.2024.1363708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
Osteoporosis after bariatric surgery is an increasing health concern as the rate of bariatric surgery has risen. In animal studies mimicking bariatric procedures, bone disease, together with decreased serum levels of Ca2+, Mg2+ and the gastric hormone Ghrelin were described. Ghrelin regulates metabolism by binding to and activating the growth hormone secretagogue receptor (GHSR) which is also expressed in the kidney. As calcium and magnesium are key components of bone, we tested the hypothesis that Ghrelin-deficiency contributes to osteoporosis via reduced upregulation of the renal calcium channel TRPV5 and the heteromeric magnesium channel TRPM6/7. We expressed GHSR with TRPV5 or TRPM6/7 channel in HEK293 cells and treated them with purified Ghrelin. Whole-cell current density was analyzed by patch-clamp recording. Nephron-specific gene expression was performed by tubular microdissection followed by qPCR in wild-type (WT) mice, and immunofluorescent imaging of GHSR-eGFP mice. Tubular magnesium homeostasis was analyzed in GHSR-null and WT mice at baseline and after caloric restriction. After Ghrelin exposure, whole-cell current density did not change for TRPV5 but increased for TRPM6/7 in a dose-dependent fashion. Applying the Ghrelin-mimetic (D-Trp7, Ala8,D-Phe10)-α-MSH (6-11) amide without and with the GHSR antagonist (D-Lys3)-GHRP6, we confirmed the stimulatory role of Ghrelin towards TRPM6/7. As GHSR initiates downstream signaling via protein kinase A (PKA), we found that the PKA inhibitor H89 abrogated TRPM6/7 stimulation by Ghrelin. Similarly, transfected Gαs, but not the Gαs mutant Q227L, nor Gαi2, Gαq, or Gα13 upregulated TRPM6/7 current density. In microdissected TALs and DCTs similar levels of GHSR mRNA were detected. In contrast, TRPM6 mRNA was expressed in the DCT and also detected in the TAL at 25% expression compared to DCT. Immunofluorescent studies using reporter GHSR-eGFP mice showed a strong eGFP signal in the TAL but surprisingly displayed no eGFP signal in the DCT. In 3-, 6-, and 9-month-old GHSR-null and WT mice, baseline serum magnesium was not significantly different, but 24-h urinary magnesium excretion was elevated in 9-month-old GHSR-null mice. In calorically restricted GHSR-null mice, we detected excess urinary magnesium excretion and reduced serum magnesium levels compared to WT mice. The kidneys from calorically restricted WT mice showed upregulated gene expression of magnesiotropic genes Hnf1b, Cldn-16, Cldn-19, Fxyd-2b, and Parvalbumin compared to GHSR-null mice. Our in vitro studies show that Ghrelin stimulates TRPM6/7 via GHSR and Gαs-PKA signaling. The murine studies are consistent with Ghrelin-GHSR signaling inducing reduced urinary magnesium excretion, particularly in calorically restricted mice when Ghrelin levels are elevated. This effect may be mediated by Ghrelin-upregulation of TRPM6 in the TAL and/or upregulation of other magnesiotropic genes. We postulate that rising Ghrelin levels with hunger contribute to increased renal Mg2+ reabsorption to compensate for lack of enteral Mg2+ uptake.
Collapse
Affiliation(s)
- Mingzhu Nie
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jing Zhang
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Manjot Bal
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Claudia Duran
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sung Wan An
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Pediatric Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - Jeffrey M. Zigman
- Department of Internal Medicine, Center for Hypothalamic Research, UTSW Medical Center, Dallas, TX, United States
| | - Michel Baum
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chitkale Hiremath
- Department of Internal Medicine, Nephrology, and Department of Cell Biology, UTSW Medical Center, Dallas, TX, United States
| | - Denise K. Marciano
- Department of Internal Medicine, Nephrology, and Department of Cell Biology, UTSW Medical Center, Dallas, TX, United States
| | - Matthias T. F. Wolf
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Pediatric Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Su XT, Reyes JV, Lackey AE, Demirci H, Bachmann S, Maeoka Y, Cornelius RJ, McCormick JA, Yang CL, Jung HJ, Welling PA, Nelson JW, Ellison DH. Enriched Single-Nucleus RNA-Sequencing Reveals Unique Attributes of Distal Convoluted Tubule Cells. J Am Soc Nephrol 2024; 35:426-440. [PMID: 38238903 PMCID: PMC11000721 DOI: 10.1681/asn.0000000000000297] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
SIGNIFICANCE STATEMENT High-resolution single-nucleus RNA-sequencing data indicate a clear separation between primary sites of calcium and magnesium handling within distal convoluted tubule (DCT). Both DCT1 and DCT2 express Slc12a3, but these subsegments serve distinctive functions, with more abundant magnesium-handling genes along DCT1 and more calcium-handling genes along DCT2. The data also provide insight into the plasticity of the distal nephron-collecting duct junction, formed from cells of separate embryonic origins. By focusing/changing gradients of gene expression, the DCT can morph into different physiological cell states on demand. BACKGROUND The distal convoluted tubule (DCT) comprises two subsegments, DCT1 and DCT2, with different functional and molecular characteristics. The functional and molecular distinction between these segments, however, has been controversial. METHODS To understand the heterogeneity within the DCT population with better clarity, we enriched for DCT nuclei by using a mouse line combining "Isolation of Nuclei Tagged in specific Cell Types" and sodium chloride cotransporter-driven inducible Cre recombinase. We sorted the fluorescently labeled DCT nuclei using Fluorescence-Activated Nucleus Sorting and performed single-nucleus transcriptomics. RESULTS Among 25,183 DCT cells, 75% were from DCT1 and 25% were from DCT2. In addition, there was a small population (<1%) enriched in proliferation-related genes, such as Top2a , Cenpp , and Mki67 . Although both DCT1 and DCT2 expressed sodium chloride cotransporter, magnesium transport genes were predominantly expressed along DCT1, whereas calcium, electrogenic sodium, and potassium transport genes were more abundant along DCT2. The transition between these two segments was gradual, with a transitional zone in which DCT1 and DCT2 cells were interspersed. The expression of the homeobox genes by DCT cells suggests that they develop along different trajectories. CONCLUSIONS Transcriptomic analysis of an enriched rare cell population using a genetically targeted approach clarifies the function and classification of distal cells. The DCT segment is short, can be separated into two subsegments that serve distinct functions, and is speculated to derive from different origins during development.
Collapse
Affiliation(s)
- Xiao-Tong Su
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Jeremiah V. Reyes
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Anne E. Lackey
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Hasan Demirci
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Bachmann
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Yujiro Maeoka
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Ryan J. Cornelius
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - James A. McCormick
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Hyun Jun Jung
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paul A. Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan W. Nelson
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - David H. Ellison
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon
- Renal Section, VA Portland Healthcare System, Portland, Oregon
| |
Collapse
|
17
|
Bosman W, Franken GAC, de Las Heras J, Madariaga L, Barakat TS, Oostenbrink R, van Slegtenhorst M, Perdomo-Ramírez A, Claverie-Martín F, van Eerde AM, Vargas-Poussou R, Dubourg LD, González-Recio I, Martínez-Cruz LA, de Baaij JHF, Hoenderop JGJ. Hypomagnesaemia with varying degrees of extrarenal symptoms as a consequence of heterozygous CNNM2 variants. Sci Rep 2024; 14:6917. [PMID: 38519529 PMCID: PMC10959950 DOI: 10.1038/s41598-024-57061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Variants in the CNNM2 gene are causative for hypomagnesaemia, seizures and intellectual disability, although the phenotypes can be variable. This study aims to understand the genotype-phenotype relationship in affected individuals with CNNM2 variants by phenotypic, functional and structural analysis of new as well as previously reported variants. This results in the identification of seven variants that significantly affect CNNM2-mediated Mg2+ transport. Pathogenicity of these variants is further supported by structural modelling, which predicts CNNM2 structure to be affected by all of them. Strikingly, seizures and intellectual disability are absent in 4 out of 7 cases, indicating these phenotypes are caused either by specific CNNM2 variant only or by additional risk factors. Moreover, in line with sporadic observations from previous reports, CNNM2 variants might be associated with disturbances in parathyroid hormone and Ca2+ homeostasis.
Collapse
Affiliation(s)
- Willem Bosman
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
| | - Gijs A C Franken
- Department of Medical BioSciences, Radboudumc, Nijmegen, The Netherlands
| | - Javier de Las Heras
- Division of Pediatric Metabolism, Cruces University Hospital, CIBER-ER, Metab-ERN, University of the Basque Country (UPV/EHU), Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Leire Madariaga
- Pediatric Nephrology Department, Cruces University Hospital, CIBERDEM, CIBER-ER, Endo-ERN, Biocruces Bizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Barakaldo, Spain
| | - Tahsin Stefan Barakat
- Deparment of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
- Discovery Unit, Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
| | - Rianne Oostenbrink
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, The Netherlands
- Department of General Pediatrics, Erasmus Medical Center Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Ana Perdomo-Ramírez
- Unidad de Investigación, Renal Tube Group, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Félix Claverie-Martín
- Unidad de Investigación, Renal Tube Group, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - Rosa Vargas-Poussou
- Service de medecine genomique des maladies rares, AP-HP, universite Paris Cité, Paris, France
- Centre de reference des maladies renales hereditaires de l'enfant et de l'adulte MARHEA, hopital Européen Georges Pompidou, Paris, France
- CNRS, centre de recherche des Cordeliers, Inserm UMRS 1138, Sorbonne universite, universite Paris Cité, Paris, France
| | - Laurence Derain Dubourg
- Hôpital Édouard Herriot, Hospices civils de Lyon, service de nephrologie, dialyse, hypertension et exploration fonctionnelle renale, Lyon, France
- Centre de reference des maladies renales rares et phosphocalciques, Nephrogones, Hôpital Femme-Mère-Enfant Bron, Bron, France
- Faculté de medecine Lyon est, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Irene González-Recio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Luis Alfonso Martínez-Cruz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | | | | |
Collapse
|
18
|
Chubanov V, Köttgen M, Touyz RM, Gudermann T. TRPM channels in health and disease. Nat Rev Nephrol 2024; 20:175-187. [PMID: 37853091 DOI: 10.1038/s41581-023-00777-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Different cell channels and transporters tightly regulate cytoplasmic levels and the intraorganelle distribution of cations. Perturbations in these processes lead to human diseases that are frequently associated with kidney impairment. The family of melastatin-related transient receptor potential (TRPM) channels, which has eight members in mammals (TRPM1-TRPM8), includes ion channels that are highly permeable to divalent cations, such as Ca2+, Mg2+ and Zn2+ (TRPM1, TRPM3, TRPM6 and TRPM7), non-selective cation channels (TRPM2 and TRPM8) and monovalent cation-selective channels (TRPM4 and TRPM5). Three family members contain an enzymatic protein moiety: TRPM6 and TRPM7 are fused to α-kinase domains, whereas TRPM2 is linked to an ADP-ribose-binding NUDT9 homology domain. TRPM channels also function as crucial cellular sensors involved in many physiological processes, including mineral homeostasis, blood pressure, cardiac rhythm and immunity, as well as photoreception, taste reception and thermoreception. TRPM channels are abundantly expressed in the kidney. Mutations in TRPM genes cause several inherited human diseases, and preclinical studies in animal models of human disease have highlighted TRPM channels as promising new therapeutic targets. Here, we provide an overview of this rapidly evolving research area and delineate the emerging role of TRPM channels in kidney pathophysiology.
Collapse
Affiliation(s)
- Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| | - Michael Köttgen
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Rhian M Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| |
Collapse
|
19
|
Roskosch J, Huynh-Do U, Rudloff S. Lectin-mediated, time-efficient, and high-yield sorting of different morphologically intact nephron segments. Pflugers Arch 2024; 476:379-393. [PMID: 38091061 PMCID: PMC10847228 DOI: 10.1007/s00424-023-02894-w] [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: 02/28/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/08/2024]
Abstract
The kidney is a highly complex organ equipped with a multitude of miniscule filter-tubule units called nephrons. Each nephron can be subdivided into multiple segments, each with its own morphology and physiological function. To date, conventional manual approaches to isolate specific nephron segments are very laborious, time-consuming, often limited to only a specific segment, and typically have low yield. Here, we describe a novel, unconventional method that is superior in many aspects to previous protocols by combining low-cost fluorophore-conjugated lectins or agglutinins (Flaggs) with flow sorting. This allows the simultaneous separation of different nephron segments with preserved 3D morphology from mouse or human samples in under 3 h. Using a 200-µm nozzle and 5 psi, glomeruli, proximal, or distal convoluted tubules are sorted with Cy3-labeled Sambucus Nigra agglutinin (SNA-Cy3), Fluorescein-labeled Lotus Tetragonolobus lectin (LTL-FITC), or Pacific Blue-labeled soybean agglutinin (SBA-PB), respectively. Connecting tubules and collecting ducts are sorted by double-positive SBA-PB and SNA-Cy3 signals, while thick ascending limb segments are characterized by the absence of any Flaggs labeling. From two mouse kidneys, this yields 37-521 ng protein/s or 0.71-16.71 ng RNA/s, depending on the specific nephron segment. The purity of sorted segments, as assessed by mRNA expression level profiling of 15 genes, is very high with a 96.1-fold median enrichment across all genes and sorted segments. In summary, our method represents a simple, straightforward, cost-effective, and widely applicable tool yielding high amounts of pure and morphologically largely intact renal tubule materials with the potential to propel nephron segment-specific research.
Collapse
Affiliation(s)
- Jessica Roskosch
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Uyen Huynh-Do
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Stefan Rudloff
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland.
| |
Collapse
|
20
|
Dutta P, Hakimi S, Layton AT. How the kidney regulates magnesium: a modelling study. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231484. [PMID: 38511086 PMCID: PMC10951724 DOI: 10.1098/rsos.231484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/23/2024] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
The kidneys are crucial for maintaining Mg2+ homeostasis. Along the proximal tubule and thick ascending limb, Mg2+ is reabsorbed paracellularly, while along the distal convoluted tubule (DCT), Mg2+ is reabsorbed transcellularly via transient receptor potential melastatin 6 (TRPM6). TRPM6 and other renal transporter expressions are regulated by sex hormones. To investigate renal Mg2 handling, we have developed sex-specific computational models of electrolyte transport along rat superficial nephron. Model simulations indicated that along the proximal tubule and thick ascending limb, Mg2+ and Na+ transport occur parallelly, but they are dissociated along the DCT. In addition, our models predicted higher paracellular Mg2+ permeability in females to attain similar cortical thick ascending limb fractional Mg2+ reabsorption in both sexes. Furthermore, DCT fractional Mg2+ reabsorption is higher in females than in males, allowing females to better fine-tune Mg2+ excretion. We validated our models by simulating the administration of three classes of diuretics. The model predicted significantly increased, marginally increased and significantly decreased Mg2+ excretions for loop, thiazide and K-sparing diuretics, respectively, aligning with experimental findings. The models can be used to conduct in silico studies on kidney adaptations to Mg2+ homeostasis alterations during conditions such as pregnancy, diabetes and chronic kidney disease.
Collapse
Affiliation(s)
- Pritha Dutta
- Department of Applied Mathematics, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
| | - Shervin Hakimi
- Department of Applied Mathematics, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
| | - Anita T. Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
- Department of Biology, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
- School of Pharmacology, University of Waterloo, Waterloo, OntarioN2L 3G1, Canada
| |
Collapse
|
21
|
Dokurel Çetin İ, Betül Gerik-Çelebi H, Demiral M, Çetin O. Hereditary hypomagnesemia with secondary hypocalcemia caused by a novel mutation in TRPM6 gene. J Pediatr Endocrinol Metab 2024; 37:184-188. [PMID: 38084506 DOI: 10.1515/jpem-2023-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/27/2023] [Indexed: 02/09/2024]
Abstract
OBJECTIVES Hereditary hypomagnesemia with secondary hypocalcemia (HSH), which results from variations in the transient receptor potential melastatin 6 (TRPM6) genes, is a rare hereditary cause of extremely low serum magnesium levels. We describe an infant with triggered seizures due to hypomagnesemia and a novel mutation in TRPM6 gene was identified. CASE PRESENTATION A 10-month-old boy presented with multidrug resistant seizures, and axial hypotonia due to severe hypomagnesemia. Electroencephalography and neuroimaging of the patient was normal. He had a favorable outcome with magnesium supplement. In this study, the patient underwent clinical exome sequencing (CES) which detected a novel homozygous variant in the TRPM6 gene: NM_017662.5: c.5571-3C>G. After replacing his magnesium orally, he was free from seizures and had an encouraging outcome at the twelfth-month follow-up. CONCLUSIONS HSH often presents with developmental issues, treatment-resistant seizures, and increased neuromuscular excitability. Untreated hypomagnesemia can potentially be fatal and severely impair cognitive function. Clinical suspicion is essential for early diagnosis and treatment.
Collapse
Affiliation(s)
- İpek Dokurel Çetin
- Department of Pediatrics, Division of Pediatric Neurology, Balikesir University Medical Faculty, Balikesir, Türkiye
| | | | - Meliha Demiral
- Department of Pediatrics, Division of Pediatric Endocrinology, Ataturk City Hospital, Balikesir, Türkiye
| | - Orkun Çetin
- Department of Obstetrics and Gynecology, Division of Perinatology, Balikesir University Medical Faculty, Balıkesir, Türkiye
| |
Collapse
|
22
|
Kumar A, Mehan S, Tiwari A, Khan Z, Gupta GD, Narula AS, Samant R. Magnesium (Mg 2+): Essential Mineral for Neuronal Health: From Cellular Biochemistry to Cognitive Health and Behavior Regulation. Curr Pharm Des 2024; 30:3074-3107. [PMID: 39253923 DOI: 10.2174/0113816128321466240816075041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 09/11/2024]
Abstract
Magnesium (Mg2+) is a crucial mineral involved in numerous cellular processes critical for neuronal health and function. This review explores the multifaceted roles of Mg2+, from its biochemical interactions at the cellular level to its impact on cognitive health and behavioral regulation. Mg2+ acts as a cofactor for over 300 enzymatic reactions, including those involved in ATP synthesis, nucleic acid stability, and neurotransmitter release. It regulates ion channels, modulates synaptic plasticity, and maintains the structural integrity of cell membranes, which are essential for proper neuronal signaling and synaptic transmission. Recent studies have highlighted the significance of Mg2+ in neuroprotection, showing its ability to attenuate oxidative stress, reduce inflammation, and mitigate excitotoxicity, thereby safeguarding neuronal health. Furthermore, Mg2+ deficiency has been linked to a range of neuropsychiatric disorders, including depression, anxiety, and cognitive decline. Supplementation with Mg2+, particularly in the form of bioavailable compounds such as Magnesium-L-Threonate (MgLT), Magnesium-Acetyl-Taurate (MgAT), and other Magnesium salts, has shown some promising results in enhancing synaptic density, improving memory function, and alleviating symptoms of mental health disorders. This review highlights significant current findings on the cellular mechanisms by which Mg2+ exerts its neuroprotective effects and evaluates clinical and preclinical evidence supporting its therapeutic potential. By elucidating the comprehensive role of Mg2+ in neuronal health, this review aims to underscore the importance of maintaining optimal Mg2+ levels for cognitive function and behavioral regulation, advocating for further research into Mg2+ supplementation as a viable intervention for neuropsychiatric and neurodegenerative conditions.
Collapse
Affiliation(s)
- Aakash Kumar
- Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India) Moga, Punjab, India
| | - Sidharth Mehan
- 1Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India) Moga, Punjab, India
| | - Aarti Tiwari
- Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India) Moga, Punjab, India
| | - Zuber Khan
- Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India) Moga, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India) Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Rajaram Samant
- Department of Research and Development, Celagenex Research, Thane, Maharashtra, India
| |
Collapse
|
23
|
Garcia-Nieto VM, Claverie-Martin F, Moraleda-Mesa T, Perdomo-Ramírez A, Fraga-Rodríguez GM, Luis-Yanes MI, Ramos-Trujillo E. Renal diseases that course with hypomagnesemia. Comments on a new hereditary hypomagnesemic tubulopathy. Nefrologia 2024; 44:23-31. [PMID: 38350738 DOI: 10.1016/j.nefroe.2024.02.003] [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: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 02/15/2024] Open
Abstract
Renal diseases associated with hypomagnesemia are a complex and diverse group of tubulopathies caused by mutations in genes encoding proteins that are expressed in the thick ascending limb of the loop of Henle and in the distal convoluted tubule. In this paper, we review the initial description, the clinical expressiveness and etiology of four of the first hypomagnesemic tubulopathies described: type 3 Bartter and Gitelman diseases, Autosomal recessive hypomagnesemia with secondary hypocalcemia and Familial hypomagnesemia with hypercalciuria and nephrocalcinosis. The basic biochemical patterns observed in renal tubular hypomagnesemias and the modalities of transport and interaction that occur between the transporters involved in the reabsorption of magnesium in the distal convoluted tubule are described below. Finally, the recent report of a new renal disease with hypomagnesemia, type 2 hypomagnesemia with secondary hypocalcemia caused by reduced TRPM7 channel activity is described.
Collapse
Affiliation(s)
- Víctor M Garcia-Nieto
- Unidad de Nefrología Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.
| | - Félix Claverie-Martin
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Teresa Moraleda-Mesa
- Unidad de Nefrología Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Ana Perdomo-Ramírez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Gloria Mª Fraga-Rodríguez
- Nefrologia Pediàtrica, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Isabel Luis-Yanes
- Unidad de Nefrología Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Elena Ramos-Trujillo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Departamento de Medicina Física y Farmacología, Facultad de Medicina, Universidad de la Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
24
|
Sarna MK, Goel P, Bhargava V, Parakh R. Fahr's syndrome associated with hypoparathyroidism: A case report. J R Coll Physicians Edinb 2023; 53:283-287. [PMID: 37936282 DOI: 10.1177/14782715231210606] [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: 11/09/2023] Open
Abstract
Fahr's syndrome affects fewer than 1 in 100,000 people. It is an inherited neurological disorder, which is distinguished by atypical calcium deposition in the movement-controlling areas of brain, that is thalamus, dentate nucleus, basal ganglia, cerebellum, cerebral cortex, hippocampus and subcortical white matter. The majority of patients often experience extrapyramidal symptoms, cerebellar signs, speech difficulty, dementia and neuropsychiatric manifestations. This disease's molecular genetics have not been thoroughly investigated. Typically, young to middle-aged adults are affected though basal ganglia calcification in hypoparathyroidism is quite uncommon. Laboratory results and radiographic brain imaging helps in reaching the diagnosis. The treatment is mainly symptomatic. We present a case of Fahr's syndrome associated with hypoparathyroidism.
Collapse
Affiliation(s)
- Mukesh Kumar Sarna
- Department of General Medicine, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Pallaavi Goel
- Mahatma Gandhi University of Medical Science & Technology, Jaipur, India
| | - Varun Bhargava
- Mahatma Gandhi University of Medical Science & Technology, Jaipur, India
| | - Rishabh Parakh
- Mahatma Gandhi University of Medical Science & Technology, Jaipur, India
| |
Collapse
|
25
|
Huang S, Ge Y, Li Y, Cui N, Tan L, Guo S, Wang S, Hao L, Lei G, Yang X. Magnesium Status, Genetic Variants of Magnesium-Related Ion Channel Transient Receptor Potential Membrane Melastatin 6 (TRPM6) and the Risk of Gestational Diabetes Mellitus in Chinese Pregnant Women: A Nested Case-Control Study. Mol Nutr Food Res 2023; 67:e2200835. [PMID: 37759402 DOI: 10.1002/mnfr.202200835] [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: 11/29/2022] [Revised: 05/10/2023] [Indexed: 09/29/2023]
Abstract
SCOPE Magnesium plays an important role in regulating glucose metabolism. The study attempts to explore association between magnesium status and single nucleotide polymorphisms (SNPs) of gene involved in magnesium absorption-transient receptor potential membrane melastatin 6 (TRPM6) and gestational diabetes mellitus (GDM) risk METHODS AND RESULTS: A nested case-control study including 170 GDM cases and matched 340 controls is conducted based on Tongji Birth Cohort. Dietary, serum, and urine magnesium are evaluated before the diagnosis of GDM. Compared to the lowest tertile, women in the highest tertile of serum magnesium are at a lower risk of GDM (adjusted odds ratio [aOR] 0.42, 95% confidence intervals [CI] 0.21-0.84). Serum magnesium is inversely associated with insulin and homeostatic model assessment of insulin resistance (β = -0.05, p = 0.002; β = -0.04, p = 0.001, respectively). The aOR for GDM in carriers of the CT or CC genotypes of TRPM6 rs2274924 compared with carriers of the TT genotype is 2.76 (95% CI 1.78-4.26). Dietary magnesium is positively associated with serum magnesium (β = 0.02, p = 0.004), but not with GDM risk. CONCLUSION Serum magnesium and the TRPM6 rs2274924 polymorphism are associated with the risk of GDM.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Yanyan Ge
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Yan Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Ningning Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Le Tan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Shu Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Shanshan Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Liping Hao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| | - Gang Lei
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street, Wuhan, Hubei, 430014, P. R. China
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, China
| |
Collapse
|
26
|
Vydra Bousova K, Zouharova M, Jiraskova K, Vetyskova V. Interaction of Calmodulin with TRPM: An Initiator of Channel Modulation. Int J Mol Sci 2023; 24:15162. [PMID: 37894842 PMCID: PMC10607381 DOI: 10.3390/ijms242015162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Transient receptor potential melastatin (TRPM) channels, a subfamily of the TRP superfamily, constitute a diverse group of ion channels involved in mediating crucial cellular processes like calcium homeostasis. These channels exhibit complex regulation, and one of the key regulatory mechanisms involves their interaction with calmodulin (CaM), a cytosol ubiquitous calcium-binding protein. The association between TRPM channels and CaM relies on the presence of specific CaM-binding domains in the channel structure. Upon CaM binding, the channel undergoes direct and/or allosteric structural changes and triggers down- or up-stream signaling pathways. According to current knowledge, ion channel members TRPM2, TRPM3, TRPM4, and TRPM6 are directly modulated by CaM, resulting in their activation or inhibition. This review specifically focuses on the interplay between TRPM channels and CaM and summarizes the current known effects of CaM interactions and modulations on TRPM channels in cellular physiology.
Collapse
|
27
|
Khajavi N, Riçku K, Schreier PCF, Gentz T, Beyerle P, Cruz E, Breit A, Reinach PS, Gudermann T. Chronic Mg 2+ Deficiency Does Not Impair Insulin Secretion in Mice. Cells 2023; 12:1790. [PMID: 37443824 PMCID: PMC10340716 DOI: 10.3390/cells12131790] [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: 06/02/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Magnesium is an essential mediator of a vast number of critical enzymatic cellular reactions in the human body. Some clinical epidemiological studies suggest that hypomagnesemia accounts for declines in insulin secretion in patients with type 2 diabetes (T2D); however, the results of various experimental studies do not support this notion. To address this discrepancy, we assessed the short- and long-term effects of hypomagnesemia on β-cell function and insulin secretion in primary mouse islets of Langerhans and in a mouse model of hypomagnesemia known as Trpm6Δ17 /fl;Villin1-Cre mice. We found that lowering the extracellular Mg2+ concentration from 1.2 mM to either 0.6 or 0.1 mM remarkably increased glucose-induced insulin secretion (GIIS) in primary islets isolated from C57BL/6 mice. Similarly, both the plasma insulin levels and GIIS rose in isolated islets of Trpm6Δ17 /fl;Villin1-Cre mice. We attribute these rises to augmented increases in intracellular Ca2+ oscillations in pancreatic β-cells. However, the glycemic metabolic profile was not impaired in Trpm6Δ17 /fl;Villin1-Cre mice, suggesting that chronic hypomagnesemia does not lead to insulin resistance. Collectively, the results of this study suggest that neither acute nor chronic Mg2+ deficiency suppresses glucose-induced rises in insulin secretion. Even though hypomagnesemia can be symptomatic of T2D, such deficiency may not account for declines in insulin release in this disease.
Collapse
Affiliation(s)
- Noushafarin Khajavi
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Klea Riçku
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Pascale C. F. Schreier
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Tanja Gentz
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Philipp Beyerle
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Emmanuel Cruz
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
| | - Peter S. Reinach
- Ophthalmology Department, Wenzhou Medical University, Wenzhou 325015, China;
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, 80539 Munich, Germany; (K.R.); (P.C.F.S.); (T.G.); (P.B.); (E.C.); (A.B.)
- German Center for Lung Research, 81377 Munich, Germany
| |
Collapse
|
28
|
Bravo M, Simón J, González-Recio I, Martinez-Cruz LA, Goikoetxea-Usandizaga N, Martínez-Chantar ML. Magnesium and Liver Metabolism Through the Lifespan. Adv Nutr 2023; 14:739-751. [PMID: 37207838 PMCID: PMC10334155 DOI: 10.1016/j.advnut.2023.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/24/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023] Open
Abstract
Within the organism, the liver is the main organ responsible for metabolic homeostasis and xenobiotic transformation. To maintain an adequate liver weight-to-bodyweight ratio, this organ has an extraordinary regenerative capacity and is able to respond to an acute insult or partial hepatectomy. Maintenance of hepatic homeostasis is crucial for the proper functioning of the liver, and in this context, adequate nutrition with macro- and micronutrient intake is mandatory. Among all known macro-minerals, magnesium has a key role in energy metabolism and in metabolic and signaling pathways that maintain liver function and physiology throughout its life span. In the present review, the cation is reported as a potential key molecule during embryogenesis, liver regeneration, and aging. The exact role of the cation during liver formation and regeneration is not fully understood due to its unclear role in the activation and inhibition of those processes, and further research in a developmental context is needed. As individuals age, they may develop hypomagnesemia, a condition that aggravates the characteristic alterations. Additionally, risk of developing liver pathologies increases with age, and hypomagnesemia may be a contributing factor. Therefore, magnesium loss must be prevented by adequate intake of magnesium-rich foods such as seeds, nuts, spinach, or rice to prevent age-related hepatic alterations and contribute to the maintenance of hepatic homeostasis. Since magnesium-rich sources include a variety of foods, a varied and balanced diet can meet both macronutrient and micronutrient needs.
Collapse
Affiliation(s)
- Miren Bravo
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain
| | - Jorge Simón
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain; Center for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Bizkaia, Spain
| | - Irene González-Recio
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain
| | - Luis Alfonso Martinez-Cruz
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain; Center for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Bizkaia, Spain.
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio (Bizkaia), Spain; Center for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Bizkaia, Spain.
| |
Collapse
|
29
|
Morrison AR. Magnesium Homeostasis: Lessons from Human Genetics. Clin J Am Soc Nephrol 2023; 18:969-978. [PMID: 36723340 PMCID: PMC10356123 DOI: 10.2215/cjn.0000000000000103] [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: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/02/2023]
Abstract
Mg 2+ , the fourth most abundant cation in the body, serves as a cofactor for about 600 cellular enzymes. One third of ingested Mg 2+ is absorbed from the gut through a saturable transcellular process and a concentration-dependent paracellular process. Absorbed Mg 2+ is excreted by the kidney and maintains serum Mg 2+ within a narrow range of 0.7-1.25 mmol/L. The reabsorption of Mg 2+ by the nephron is characterized by paracellular transport in the proximal tubule and thick ascending limb. The nature of the transport pathways in the gut epithelia and thick ascending limb has emerged from an understanding of the molecular mechanisms responsible for rare monogenetic disorders presenting with clinical hypomagnesemia. These human disorders due to loss-of-function mutations, in concert with mouse models, have led to a deeper understanding of Mg 2+ transport in the gut and renal tubule. This review focuses on the nature of the transporters and channels revealed by human and mouse genetics and how they are integrated into an understanding of human Mg 2+ physiology.
Collapse
Affiliation(s)
- Aubrey R Morrison
- Division of Nephrology, Department of Medicine and Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
30
|
Aal-Hamad AH, Al-Alawi AM, Kashoub MS, Falhammar H. Hypermagnesemia in Clinical Practice. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1190. [PMID: 37512002 PMCID: PMC10384947 DOI: 10.3390/medicina59071190] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Hypermagnesemia is a relatively uncommon but potentially life-threatening electrolyte disturbance characterized by elevated magnesium concentrations in the blood. Magnesium is a crucial mineral involved in various physiological functions, such as neuromuscular conduction, cardiac excitability, vasomotor tone, insulin metabolism, and muscular contraction. Hypomagnesemia is a prevalent electrolyte disturbance that can lead to several neuromuscular, cardiac, or nervous system disorders. Hypermagnesemia has been associated with adverse clinical outcomes, particularly in hospitalized patients. Prompt identification and management of hypermagnesemia are crucial to prevent complications, such as respiratory and cardiovascular negative outcomes, neuromuscular dysfunction, and coma. Preventing hypermagnesemia is crucial, particularly in high-risk populations, such as patients with impaired renal function or those receiving magnesium-containing medications or supplements. Clinical management of hypermagnesemia involves discontinuing magnesium-containing therapies, intravenous fluid therapy, or dialysis in severe cases. Furthermore, healthcare providers should monitor serum magnesium concentration in patients at risk of hypermagnesemia and promptly intervene if the concentration exceeds the normal range.
Collapse
Affiliation(s)
- Aya Hasan Aal-Hamad
- Department of Emergency, Sultan Qaboos University Hospital, P.O. Box 141, Muscat 123, Oman
| | - Abdullah M Al-Alawi
- Department of Medicine, Sultan Qaboos University Hospital, P.O. Box 141, Muscat 123, Oman
- Internal Medicine Residency Training Program, Oman Medical Specialty Board, P.O. Box 1422, Al-Khoudh 132, Oman
| | - Masoud Salim Kashoub
- Department of Medicine, Sultan Qaboos University Hospital, P.O. Box 141, Muscat 123, Oman
- Internal Medicine Residency Training Program, Oman Medical Specialty Board, P.O. Box 1422, Al-Khoudh 132, Oman
| | - Henrik Falhammar
- Department of Endocrinology, QB85, Karolinska University Hospital, 17176 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, 17177 Stockholm, Sweden
| |
Collapse
|
31
|
Gelineau-van Waes J, van Waes MA, Hallgren J, Hulen J, Bredehoeft M, Ashley-Koch AE, Krupp D, Gregory SG, Stessman HA. Gene-nutrient interactions that impact magnesium homeostasis increase risk for neural tube defects in mice exposed to dolutegravir. Front Cell Dev Biol 2023; 11:1175917. [PMID: 37377737 PMCID: PMC10292217 DOI: 10.3389/fcell.2023.1175917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
In 2018, data from a surveillance study in Botswana evaluating adverse birth outcomes raised concerns that women on antiretroviral therapy (ART) containing dolutegravir (DTG) may be at increased risk for neural tube defects (NTDs). The mechanism of action for DTG involves chelation of Mg2+ ions in the active site of the viral integrase. Plasma Mg2+ homeostasis is maintained primarily through dietary intake and reabsorption in the kidneys. Inadequate dietary Mg2+ intake over several months results in slow depletion of plasma Mg2+ and chronic latent hypomagnesemia, a condition prevalent in women of reproductive age worldwide. Mg2+ is critical for normal embryonic development and neural tube closure. We hypothesized that DTG therapy might slowly deplete plasma Mg2+ and reduce the amount available to the embryo, and that mice with pre-existing hypomagnesemia due to genetic variation and/or dietary Mg2+ insufficiency at the time of conception and initiation of DTG treatment would be at increased risk for NTDs. We used two different approaches to test our hypothesis: 1) we selected mouse strains that had inherently different basal plasma Mg2+ levels and 2) placed mice on diets with different concentrations of Mg2+. Plasma and urine Mg2+ were determined prior to timed mating. Pregnant mice were treated daily with vehicle or DTG beginning on the day of conception and embryos examined for NTDs on gestational day 9.5. Plasma DTG was measured for pharmacokinetic analysis. Our results demonstrate that hypomagnesemia prior to conception, due to genetic variation and/or insufficient dietary Mg2+ intake, increases the risk for NTDs in mice exposed to DTG. We also analyzed whole-exome sequencing data from inbred mouse strains and identified 9 predicted deleterious missense variants in Fam111a that were unique to the LM/Bc strain. Human FAM111A variants are associated with hypomagnesemia and renal Mg2+ wasting. The LM/Bc strain exhibits this same phenotype and was the strain most susceptible to DTG-NTDs. Our results suggest that monitoring plasma Mg2+ levels in patients on ART regimens that include DTG, identifying other risk factors that impact Mg2+ homeostasis, and correcting deficiencies in this micronutrient might provide an effective strategy for mitigating NTD risk.
Collapse
Affiliation(s)
- J. Gelineau-van Waes
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | | | - J. Hallgren
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - J. Hulen
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - M. Bredehoeft
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - A. E. Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - D. Krupp
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - S. G. Gregory
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - H. A. Stessman
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| |
Collapse
|
32
|
De Falco A, Iolascon A, Ascione F, Piscopo C. New Insights in 9q21.13 Microdeletion Syndrome: Genotype-Phenotype Correlation of 28 Patients. Genes (Basel) 2023; 14:genes14051116. [PMID: 37239476 DOI: 10.3390/genes14051116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The implementation of array comparative genomic hybridisation (array-CGH) allows us to describe new microdeletion/microduplication syndromes which were previously not identified. 9q21.13 microdeletion syndrome is a genetic condition due to the loss of a critical genomic region of approximately 750kb and includes several genes, such as RORB and TRPM6. Here, we report a case of a 7-year-old boy affected by 9q21.13 microdeletion syndrome. He presents with global developmental delay, intellectual disability, autistic behaviour, seizures and facial dysmorphism. Moreover, he has severe myopia, which was previously reported in only another patient with 9q21.13 deletion, and brain anomalies which were never described before in 9q21.13 microdeletion syndrome. We also collect 17 patients from a literature search and 10 cases from DECIPHER database with a total number of 28 patients (including our case). In order to better investigate the four candidate genes RORB, TRPM6, PCSK5, and PRUNE2 for neurological phenotype, we make, for the first time, a classification in four groups of all the collected 28 patients. This classification is based both on the genomic position of the deletions included in the 9q21.3 locus deleted in our patient and on the different involvement of the four-candidate gene. In this way, we compare the clinical problems, the radiological findings, and the dysmorphic features of each group and of all the 28 patients in our article. Moreover, we perform the genotype-phenotype correlation of the 28 patients to better define the syndromic spectrum of 9q21.13 microdeletion syndrome. Finally, we propose a baseline ophthalmological and neurological monitoring of this syndrome.
Collapse
Affiliation(s)
- Alessandro De Falco
- U.O.C. Genetica Medica, A.O.U. Federico II, 80131 Naples, Italy
- Dipartimento di Medicina Molecolare di Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy
| | - Achille Iolascon
- U.O.C. Genetica Medica, A.O.U. Federico II, 80131 Naples, Italy
- Dipartimento di Medicina Molecolare di Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
| | - Flora Ascione
- Hospital Directorate, A.O.R.N. "Antonio Cardarelli", 80100 Naples, Italy
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. "Antonio Cardarelli", 80100 Naples, Italy
| |
Collapse
|
33
|
Uthayabalan S, Vishnu N, Madesh M, Stathopulos PB. The human MRS2 magnesium-binding domain is a regulatory feedback switch for channel activity. Life Sci Alliance 2023; 6:e202201742. [PMID: 36754568 PMCID: PMC9909464 DOI: 10.26508/lsa.202201742] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Mitochondrial RNA splicing 2 (MRS2) forms a magnesium (Mg2+) entry protein channel in mitochondria. Whereas MRS2 contains two transmembrane domains constituting a pore on the inner mitochondrial membrane, most of the protein resides within the matrix. Yet, the precise structural and functional role of this obtrusive amino terminal domain (NTD) in human MRS2 is unknown. Here, we show that the MRS2 NTD self-associates into a homodimer, contrasting the pentameric assembly of CorA, an orthologous bacterial channel. Mg2+ and calcium suppress lower and higher order oligomerization of MRS2 NTD, whereas cobalt has no effect on the NTD but disassembles full-length MRS2. Mutating-pinpointed residues-mediating Mg2+ binding to the NTD not only selectively decreases Mg2+-binding affinity ∼sevenfold but also abrogates Mg2+ binding-induced secondary, tertiary, and quaternary structure changes. Disruption of NTD Mg2+ binding strikingly potentiates mitochondrial Mg2+ uptake in WT and Mrs2 knockout cells. Our work exposes a mechanism for human MRS2 autoregulation by negative feedback from the NTD and identifies a novel gain of function mutant with broad applicability to future Mg2+ signaling research.
Collapse
Affiliation(s)
- Sukanthathulse Uthayabalan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Neelanjan Vishnu
- Center for Mitochondrial Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Muniswamy Madesh
- Center for Mitochondrial Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| |
Collapse
|
34
|
Ray E, Mohan K, Ahmad S, Wolf MTF. Physiology of a Forgotten Electrolyte-Magnesium Disorders. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:148-163. [PMID: 36868730 DOI: 10.1053/j.akdh.2022.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/26/2022] [Accepted: 12/06/2022] [Indexed: 03/05/2023]
Abstract
Magnesium (Mg2+) is the second most common intracellular cation and the fourth most abundant element on earth. However, Mg2+ is a frequently overlooked electrolyte and often not measured in patients. While hypomagnesemia is common in 15% of the general population, hypermagnesemia is typically only found in preeclamptic women after Mg2+ therapy and in patients with ESRD. Mild to moderate hypomagnesemia has been associated with hypertension, metabolic syndrome, type 2 diabetes mellitus, CKD, and cancer. Nutritional Mg2+ intake and enteral Mg2+ absorption are important for Mg2+ homeostasis, but the kidneys are the key regulators of Mg2+ homeostasis by limiting urinary excretion to less than 4% while the gastrointestinal tract loses over 50% of the Mg2+ intake in the feces. Here, we review the physiological relevance of Mg2+, the current knowledge of Mg2+ absorption in the kidneys and the gut, the different causes of hypomagnesemia, and a diagnostic approach on how to assess Mg2+ status. We highlight the latest discoveries of monogenetic conditions causing hypomagnesemia, which have enhanced our understanding of tubular Mg2+ absorption. We will also discuss external and iatrogenic causes of hypomagnesemia and advances in the treatment of hypomagnesemia.
Collapse
Affiliation(s)
- Evan Ray
- Renal-Electrolyte Division, Department of Internal Medicine, University of Pittsburgh, PA
| | - Krithika Mohan
- Department of Nephrology, Hosmat Hospital, HBR Layout, Bangalore, India
| | - Syeda Ahmad
- Renal-Electrolyte Division, Department of Internal Medicine, University of Pittsburgh, PA
| | - Matthias T F Wolf
- Pediatric Nephrology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
35
|
Abstract
Mg2+ is essential for many cellular and physiological processes, including muscle contraction, neuronal activity, and metabolism. Consequently, the blood Mg2+ concentration is tightly regulated by balanced intestinal Mg2+ absorption, renal Mg2+ excretion, and Mg2+ storage in bone and soft tissues. In recent years, the development of novel transgenic animal models and identification of Mendelian disorders has advanced our current insight in the molecular mechanisms of Mg2+ reabsorption in the kidney. In the proximal tubule, Mg2+ reabsorption is dependent on paracellular permeability by claudin-2/12. In the thick ascending limb of Henle's loop, the claudin-16/19 complex provides a cation-selective pore for paracellular Mg2+ reabsorption. The paracellular Mg2+ reabsorption in this segment is regulated by the Ca2+-sensing receptor, parathyroid hormone, and mechanistic target of rapamycin (mTOR) signaling. In the distal convoluted tubule, the fine tuning of Mg2+ reabsorption takes place by transcellular Mg2+ reabsorption via transient receptor potential melastatin-like types 6 and 7 (TRPM6/TRPM7) divalent cation channels. Activity of TRPM6/TRPM7 is dependent on hormonal regulation, metabolic activity, and interacting proteins. Basolateral Mg2+ extrusion is still poorly understood but is probably dependent on the Na+ gradient. Cyclin M2 and SLC41A3 are the main candidates to act as Na+/Mg2+ exchangers. Consequently, disturbances of basolateral Na+/K+ transport indirectly result in impaired renal Mg2+ reabsorption in the distal convoluted tubule. Altogether, this review aims to provide an overview of the molecular mechanisms of Mg2+ reabsorption in the kidney, specifically focusing on transgenic mouse models and human hereditary diseases.
Collapse
Affiliation(s)
- Jeroen H F de Baaij
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Mirbod SM, Khanahmad H, Amerizadeh A, Amirpour A, Mirbod SM, Zaker E. Viewpoints on the Role of Transient Receptor Potential Melastatin Channels in Cardiovascular System and Disease: A Systematic Review. Curr Probl Cardiol 2023; 48:101012. [PMID: 34644560 DOI: 10.1016/j.cpcardiol.2021.101012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/04/2023]
Abstract
Transient receptor potential (TRP) family play critical roles in cardiovascular system. TRPM family as largest TRP subfamily is non-voltage Ca2+-activated selective channels which has 8 members. This study aimed to discuss the role of TRPM family in cardiovascular system and diseases. Systematic search was performed covering PubMed, ISI Web of Science, and Google Scholar from inception until June 2021 using related keywords and Mesh terms for English studies with human, animal and in-vitro subjects. Finally 10 studies were selected for data extraction. Reviewing the articles showed that TRPM2, TRPM4, TRPM5, TRPM6 and TRPM7 play important roles in cardiovascular system and diseases. TRPM2 could be activated by reactive oxygen species (ROS) and effects on cardiac injury and cardiac fibrosis. TRPM7 and TRPM6 also have been reported to be associated with cardiac fibrosis and atrial fibrosis development respectively. TRPM4 channels contributed to resting membrane potential of cerebral artery smooth muscle cells and atrial contraction. TRPM5 channels are bitter taste sensors and prevent high salt intake and consequently high blood pressure due to the high salt intake. In conclusion based on the proof of the effectiveness of some members of TRPM family in the cardiovascular system, research on other members of this channel group seems to be useful and necessary to find their possible connection to the cardiovascular system.
Collapse
Affiliation(s)
| | - Hossein Khanahmad
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Atefeh Amerizadeh
- Department of Cardiology, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Department of Physiology, Cardiovascular Research Institute, Isfahan University of Medical sciences, Isfahan, Iran
| | - Afshin Amirpour
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Mojgan Mirbod
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Zaker
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
37
|
Chamniansawat S, Suksridechacin N, Thongon N. Current opinion on the regulation of small intestinal magnesium absorption. World J Gastroenterol 2023; 29:332-342. [PMID: 36687126 PMCID: PMC9846944 DOI: 10.3748/wjg.v29.i2.332] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/25/2022] [Accepted: 11/19/2022] [Indexed: 01/06/2023] Open
Abstract
Magnesium (Mg2+) has an important role in numerous biological functions, and Mg2+ deficiency is associated with several diseases. Therefore, adequate intestinal absorption of Mg2+ is vital for health. The small intestine was previously thought to absorb digested Mg2+ exclusively through an unregulated paracellular mechanism, which is responsible for approximately 90% of total Mg2+ absorption. Recent studies, however, have revealed that the duodenum, jejunum, and ileum absorb Mg2+ through both transcellular and paracellular routes. Several regulatory factors of small intestinal Mg2+ uptake also have been explored, e.g., parathyroid hormone, fibroblast growth factor-23, apical acidity, proton pump inhibitor, and pH-sensing channel and receptors. The mechanistic factors underlying proton pump inhibitor suppression of small intestinal Mg2+, such as magnesiotropic protein dysfunction, higher mucosal bicarbonate secretion, Paneth cell dysfunction, and intestinal inflammation, are currently being explored. The potential role of small intestinal microbiomes in Mg2+ absorption has also been proposed. In this article, we reviewed the current knowledge on the mechanisms and regulatory factors of small intestinal Mg2+ absorption.
Collapse
Affiliation(s)
- Siriporn Chamniansawat
- Division of Anatomy, Department of Medical Sciences, Faculty of Allied Health Sciences, Burapha University, Muang 20131, Chonburi, Thailand
| | - Nasisorn Suksridechacin
- Biodiversity Research Centre, Thailand Institute of Scientific and Technological Research, Khlong Luang 12120, Pathum Thani, Thailand
| | - Narongrit Thongon
- Division of Physiology, Department of Medical Sciences, Faculty of Allied Health Sciences, Burapha University, Muang 20131, Chonburi, Thailand
| |
Collapse
|
38
|
Romero JR, Inostroza‐Nieves Y, Pulido‐Perez P, Lopez P, Wohlgemuth JG, Dlott JS, Snyder LM, Alper SL, Rivera A. Magnesium homeostasis in deoxygenated sickle erythrocytes is modulated by endothelin-1 via Na + /Mg 2+ exchange. FASEB J 2022; 36:e22638. [PMID: 36331552 PMCID: PMC9703344 DOI: 10.1096/fj.202201339r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Painful crises in sickle cell disease (SCD) are associated with increased plasma cytokines levels, including endothelin-1 (ET-1). Reduced red cell magnesium content, mediated in part by increased Na+ /Mg2+ exchanger (NME) activity, contributes to erythrocyte K+ loss, dehydration and sickling in SCD. However, the relationship between ET-1 and the NME in SCD has remained unexamined. We observed increased NME activity in sickle red cells incubated in the presence of 500 nM ET-1. Deoxygenation of sickle red cells, in contrast, led to decreased red cell NME activity and cellular dehydration that was reversed by the NME inhibitor, imipramine. Increased NME activity in sickle red cells was significantly blocked by pre-incubation with 100 nM BQ788, a selective blocker of ET-1 type B receptors. These results suggest an important role for ET-1 and for cellular magnesium homeostasis in SCD. Consistent with these results, we observed increased NME activity in sickle red cells of three mouse models of sickle cell disease greater than that in red cells of C57BL/J6 mice. In vivo treatment of BERK sickle transgenic mice with ET-1 receptor antagonists reduced red cell NME activity. Our results suggest that ET-1 receptor blockade may be a promising therapeutic approach to control erythrocyte volume and magnesium homeostasis in SCD and may thus attenuate or retard the associated chronic inflammatory and vascular complications of SCD.
Collapse
Affiliation(s)
- José R. Romero
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Yaritza Inostroza‐Nieves
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA,Department of Biochemistry and PharmacologySan Juan Bautista School of MedicineCaguasPuerto RicoUSA
| | - Patricia Pulido‐Perez
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Pablo Lopez
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | | | | | | | - Seth L. Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Alicia Rivera
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, and Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA,Division of Laboratory Medicine, Boston Children's Hospital, and Department of PathologyHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
39
|
Andriulė I, Pangonytė D, Gwanyanya A, Karčiauskas D, Mubagwa K, Mačianskienė R. Detection of TRPM6 and TRPM7 Proteins in Normal and Diseased Cardiac Atrial Tissue and Isolated Cardiomyocytes. Int J Mol Sci 2022; 23:ijms232314860. [PMID: 36499188 PMCID: PMC9736228 DOI: 10.3390/ijms232314860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Magnesium-sensitive transient receptor potential melastatin (TRPM) ion channels, TRPM6 and TRPM7, are present in several organs, but their roles in the heart remain unclear. Therefore, here, we studied the expression patterns of TRPM6 and TRPM7 in normal and diseased myocardium. Cardiac atrial tissue and cardiomyocytes were obtained from healthy pigs and undiseased human hearts as well as from hearts of patients with ischemic heart disease (IHD) or atrial fibrillation (AF). Immunofluorescence and ELISA were used to detect TRP proteins. TRPM6 and TRPM7 immunofluorescence signals, localized at/near the cell surface or intracellularly, were detected in pig and human atrial tissues. The TRP channel modulators carvacrol (CAR, 100 µM) or 2-aminoethoxydiphenyl borate (2-APB, 500 µM) decreased the TRPM7 signal, but enhanced that of TRPM6. At a higher concentration (2 mM), 2-APB enhanced the signals of both proteins. TRPM6 and TRPM7 immunofluorescence signals and protein concentrations were increased in atrial cells and tissues from IHD or AF patients. TRPM6 and TRPM7 proteins were both detected in cardiac atrial tissue, with relatively similar subcellular localization, but distinctive drug sensitivity profiles. Their upregulated expression in IHD and AF suggests a possible role of the channels in cardiac atrial disease.
Collapse
Affiliation(s)
- Inga Andriulė
- Institute of Cardiology, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
| | - Dalia Pangonytė
- Institute of Cardiology, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
| | - Asfree Gwanyanya
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dainius Karčiauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
- Department of Cardiac, Thoracic and Vascular Surgery, Hospital of Lithuanian University of Health Sciences Kauno Klinikos, 50161 Kaunas, Lithuania
| | - Kanigula Mubagwa
- Department of Cardiovascular Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
- Department of Basic Sciences, Faculty of Medicine, Université Catholique de Bukavu, Bukavu, Congo
| | - Regina Mačianskienė
- Institute of Cardiology, Lithuanian University of Health Sciences, 50103 Kaunas, Lithuania
- Correspondence:
| |
Collapse
|
40
|
Ilenwabor BP, Franken GAC, Sponder G, Bos C, Racay P, Kolisek M, Hoenderop JGJ, de Baaij JHF. SLC41A1 knockout mice display normal magnesium homeostasis. Am J Physiol Renal Physiol 2022; 323:F553-F563. [PMID: 36049064 DOI: 10.1152/ajprenal.00101.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transcellular Mg2+ reabsorption in the distal convoluted tubule (DCT) of the kidneys plays an important role in maintaining systemic Mg2+ homeostasis. SLC41A1 is a Na+/Mg2+ exchanger that mediates Mg2+ efflux from cells and is hypothesized to facilitate basolateral extrusion of Mg2+ in the DCT. In this study, we generated a SLC41A1 knockout mouse model to examine the role of SLC41A1 in Mg2+ homeostasis. Slc41a1-/- mice exhibited similar serum and urine Mg2+ levels as their wild-type littermates. Dietary restriction of Mg2+ resulted in reduced serum Mg2+ concentration and urinary Mg2+ excretion, which was similar in the wild-type and knockout groups. Expression of genes encoding Mg2+ channels and transporters such as transient receptor potential melastatin 6 (Trpm6), transient receptor potential melastatin 7 (Trpm7), cyclin and CBS domain divalent metal cation transport mediator 2 (Cnnm2), and Slc41a3 were unchanged based on genotype. We investigated the potential redundancy of SLC41A1 and its homolog SLC41A3 by generating a double knockout mouse. Although Slc41a3-/- knockout mice showed significantly reduced serum Mg2+ compared with wild-type and Slc41a1-/- knockout groups, double knockout mice displayed similar serum Mg2+ levels as Slc41a3-/- knockout mice. In conclusion, our data show that SLC41A1 is not involved in the regulation of systemic Mg2+ homeostasis in mice. Our data also demonstrate that SLC41A1 does not compensate for the loss of SLC41A3, suggesting different functions of these SLC41 proteins in vivo.NEW & NOTEWORTHY SLC41A1 has been hypothesized to mediate Mg2+ extrusion in the distal convoluted tubule and thus regulate Mg2+ homeostasis. This study investigated the role of SLC41A1 in Mg2+ homeostasis in vivo using a transgenic mouse model. Our results demonstrate that SLC41A1 is not required to maintain normal Mg2+ balance in mice. We also show that SLC41A3 is more important than SLC41A1 in regulating systemic Mg2+ levels.
Collapse
Affiliation(s)
- Barnabas P Ilenwabor
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gijs A C Franken
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerhard Sponder
- Institute of Veterinary Physiology, Freie Universität Berlin, Berlin, Germany
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Racay
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia.,Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
41
|
Xiang X, Ji Z, Jiang T, Huang Z, Yan J. Reduced serum magnesium is associated with the occurrence of diabetic macular edema in patients with diabetic retinopathy: A retrospective study. Front Med (Lausanne) 2022; 9:923282. [PMID: 36203780 PMCID: PMC9530391 DOI: 10.3389/fmed.2022.923282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Serum magnesium levels have been reported to reflect the risk of diabetic retinopathy (DR); however, the effect of serum magnesium level on diabetic macular edema (DME) remains unclear. Here, we investigated the association between the serum magnesium levels and DME in patients with DR. Patients with DR were recruited between January 2018 and June 2021. A total of 519 such patients were included in this study. All patients underwent a standardized clinical ophthalmic examination by an experienced ophthalmologist, and an assay was conducted to determine the serum magnesium concentration. Compared with the non-DME group, the DME group had a higher proportion of insulin use and a higher level of serum ischemia-modified albumin and fasting plasma glucose. The serum magnesium and calcium levels were lower in the DME group than in the non-DME group (P < 0.05). Higher magnesium levels were negatively associated with DME after adjustment for relevant covariates. Compared with the participants in the lowest magnesium quartile, those in the fourth quartile showed a significantly lower risk of DME after adjustment [odds ratio (OR), 0.294; 95% confidence interval, 0.153–0.566; P < 0.0001]. Considering the potentially different effects of serum magnesium on the development of DME in patients with DR based on age, DR staging and insulin use, stratified analysis was performed by considering these factors. Among insulin-using patients with non-proliferative DR who were < 66 years of age, those in the third and fourth quartile of serum magnesium were less likely to develop DME than those in the lowest quartile of serum magnesium [OR (95% CI), 0.095 (0.014–0.620), 0.057 (0.011–0.305); P = 0.014, 0.001]. Overall, a higher serum magnesium level was associated with a lower risk of DME in patients with DR. Furthermore, patients with DR who used insulin were more likely to develop DME. Long-term studies on oral magnesium supplements are needed to determine whether maintaining the serum magnesium levels in a higher physiological range can reduce the risk of DME in patients with DR.
Collapse
Affiliation(s)
- Xiaoli Xiang
- Department of Ophthalmology, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, China
| | - Zijia Ji
- Department of Ultrasonography, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, China
| | - Tingwang Jiang
- Department of Key Laboratory, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, China
| | - Zhengru Huang
- Department of Ophthalmology, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, China
- *Correspondence: Zhengru Huang,
| | - Jing Yan
- School of Health Service Management, Anhui Medical University, Hefei, China
- Jing Yan,
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Gitelman syndrome is a recessive salt-wasting disorder characterized by hypomagnesemia, hypokalemia, metabolic alkalosis and hypocalciuria. The majority of patients are explained by mutations and deletions in the SLC12A3 gene, encoding the Na+-Cl--co-transporter (NCC). Recently, additional genetic causes of Gitelman-like syndromes have been identified that should be considered in genetic screening. This review aims to provide a comprehensive overview of the clinical, genetic and mechanistic aspects of Gitelman(-like) syndromes. RECENT FINDINGS Disturbed Na+ reabsorption in the distal convoluted tubule (DCT) is associated with hypomagnesemia and hypokalemic alkalosis. In Gitelman syndrome, loss-of-function mutations in SLC12A3 cause impaired NCC-mediated Na+ reabsorption. In addition, patients with mutations in CLCKNB, KCNJ10, FXYD2 or HNF1B may present with a similar phenotype, as these mutations indirectly reduce NCC activity. Furthermore, genetic investigations of patients with Na+-wasting tubulopathy have resulted in the identification of pathogenic variants in MT-TI, MT-TF, KCNJ16 and ATP1A1. These novel findings highlight the importance of cell metabolism and basolateral membrane potential for Na+ reabsorption in the DCT. SUMMARY Altogether, these findings extend the genetic spectrum of Gitelman-like electrolyte alterations. Genetic testing of patients with hypomagnesemia and hypokalemia should cover a panel of genes involved in Gitelman-like syndromes, including the mitochondrial genome.
Collapse
Affiliation(s)
- Karl P Schlingmann
- Department of General Pediatrics, University Children's Hospital, Münster, Germany
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
43
|
Kampuang N, Thongon N. Mass spectrometric analysis of TRPM6 and TRPM7 from small intestine of omeprazole-induced hypomagnesemic rats. Front Oncol 2022; 12:947899. [PMID: 36110961 PMCID: PMC9468766 DOI: 10.3389/fonc.2022.947899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Disruption of small intestinal Mg2+ absorption has been reported as the underlying mechanism of proton pump inhibitor-induced hypomagnesemia (PPIH); hence, this study evaluated the expression, localization, phosphorylation, and oxidation of transient receptor potential melastatin 6 (TRPM6) and TRPM7 in the small intestine of rats subjected to PPIH. The expression and localization of cyclin M4 (CNNM4) was also analyzed. We show that, compared to control rats, membrane expression of the TRPM6/7 heterodimer and TRPM7 was markedly lower in the duodenum and the jejunum of PPIH rats; in contrast, expression of membrane TRPM6 and CNNM4 was higher in these organs. Mass spectrometric analysis of TRPM6 demonstrated hyper-phosphorylation, especially T1851, and hyper-oxidation at M1755, both of which can suppress its channel permeability. Further, hypo-phosphorylation of S141 and the dimerization motif domain of TRPM6 in PPIH rats might be involved in lower TRPM6/7 heterodimer expression. Hypo-phosphorylation, especially at S138 and S1360 in TRPM7 from PPIH rats disrupted stability of TRPM7 at the cell membrane; hyper-oxidation of TRPM7 was also observed. These results help explain the mechanism underlying the disruption of small intestinal Mg2+ absorption in PPIH.
Collapse
|
44
|
Rössig A, Hill K, Nörenberg W, Weidenbach S, Zierler S, Schaefer M, Gudermann T, Chubanov V. Pharmacological agents selectively acting on the channel moieties of TRPM6 and TRPM7. Cell Calcium 2022; 106:102640. [PMID: 36030694 DOI: 10.1016/j.ceca.2022.102640] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022]
Abstract
The transient receptor potential cation channel, subfamily M, members 6 and 7 (TRPM6 and TRPM7) are homologous membrane proteins encompassing cation channel units fused to cytosolic serine/threonine-protein kinase domains. Clinical studies and experiments with animal disease models suggested that selective inhibition of TRPM6 and TRPM7 currents might be beneficial for subjects with immune and cardiovascular disorders, tumours and other pathologies, but the suitable pharmacological toolkit remains underdeveloped. The present study identified small synthetic molecules acting specifically on the channel moieties of TRPM6 and TRPM7. Using electrophysiological analysis in conjunction with Ca2+ imaging, we show that iloperidone and ifenprodil inhibit the channel activity of recombinant TRPM6 with IC50 values of 0.73 and 3.33 µM, respectively, without an impact on the TRPM7 channel. We also found that VER155008 suppresses the TRPM7 channel with an IC50 value of 0.11 µM but does not affect TRPM6. Finally, the effects of iloperidone and VER155008 were found to be suitable for blocking native endogenous TRPM6 and TRPM7 in a collection of mouse and human cell models. Hence, the identification of iloperidone, ifenprodil, and VER155008 allows for the first time to selectively manipulate TRPM6 and TRPM7 currents.
Collapse
Affiliation(s)
- Anna Rössig
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Kerstin Hill
- Rudolf-Boehm Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Wolfgang Nörenberg
- Rudolf-Boehm Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Sebastian Weidenbach
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Susanna Zierler
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany; Institute of Pharmacology, Johannes Kepler University Linz, Linz, Austria
| | - Michael Schaefer
- Rudolf-Boehm Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany; Comprehensive Pneumology Center, a member of the German Center for Lung Research (DZL), Munich, Germany.
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.
| |
Collapse
|
45
|
Jin F, Huang Y, Hattori M. Recent Advances in the Structural Biology of Mg 2+ Channels and Transporters. J Mol Biol 2022; 434:167729. [PMID: 35841930 DOI: 10.1016/j.jmb.2022.167729] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 10/17/2022]
Abstract
Magnesium ions (Mg2+) are the most abundant divalent cations in living organisms and are essential for various physiological processes, including ATP utilization and the catalytic activity of numerous enzymes. Therefore, the homeostatic mechanisms associated with cellular Mg2+ are crucial for both eukaryotic and prokaryotic organisms and are thus strictly controlled by Mg2+ channels and transporters. Technological advances in structural biology, such as the expression screening of membrane proteins, in meso phase crystallization, and recent cryo-EM techniques, have enabled the structure determination of numerous Mg2+ channels and transporters. In this review article, we provide an overview of the families of Mg2+ channels and transporters (MgtE/SLC41, TRPM6/7, CorA/Mrs2, CorC/CNNM), and discuss the structural biology prospects based on the known structures of MgtE, TRPM7, CorA and CorC.
Collapse
Affiliation(s)
- Fei Jin
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yichen Huang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
46
|
Khalkhali-Evrigh R, Hedayat N, Ming L, Jirimutu. Identification of selection signatures in Iranian dromedary and Bactrian camels using whole genome sequencing data. Sci Rep 2022; 12:9653. [PMID: 35688969 PMCID: PMC9187634 DOI: 10.1038/s41598-022-14376-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
The Old World camels play an important role as one of the main food sources in large parts of Asia and Africa. Natural selection combined with artificial selection by human has affected parts of the domestic animal genome for adapting them to their habitats and meeting human needs. Here, we used whole genome sequencing data of 34 camels (including 14 dromedaries and 20 Bactrian camels) to identify the genomic signature of selection in the Iranian dromedary (ID) and Bactrian camels (IB). To detect the mentioned regions, we used two methods including population differentiation index (Fst) and cross-population extended haplotype homozygosity (XP-EHH) with 50 kb sliding window and 25 kb step size. Based on gene ontology analysis on the candidate genes identified for IB camels, we found GO terms associated with lung development, nervous system development, immune system and behavior. Also, we identified several genes related to body thermoregulation (ZNF516), meat quality (ANK1 and HSPA13), and high-altitude adaptation (OPA1) for IB camels. In the list of detected candidate genes under selection in ID camels, the genes related to energy metabolism (BDH1), reproduction (DLG1, IMMP2L and FRASI), long-term memory (GRIA1), kidney (SLC12A1), lung development (EMILIN2 and FBN1) and immunity (SOCS2, JAK1, NRROS and SENP1) were found. Our findings, along with further studies in this field, will strengthen our knowledge about the effect of selection on the camelid genome under different geographical, climatic and even cultural conditions.
Collapse
Affiliation(s)
- Reza Khalkhali-Evrigh
- Department of Animal Science, Faculty of Agriculture and Natural Recourses, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Nemat Hedayat
- Department of Animal Science, Faculty of Agriculture and Natural Recourses, University of Mohaghegh Ardabili, Ardabil, Iran.
| | - Liang Ming
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot, China
| | - Jirimutu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot, China
| |
Collapse
|
47
|
Vargas-Poussou R, Claverie-Martin F, Prot-Bertoye C, Carotti V, van der Wijst J, Perdomo-Ramirez A, Fraga-Rodriguez GM, Hureaux M, Bos C, Latta F, Houillier P, Hoenderop JGJ, de Baaij JHF. Possible role for rare TRPM7 variants in patients with hypomagnesaemia with secondary hypocalcaemia. Nephrol Dial Transplant 2022; 38:679-690. [PMID: 35561741 PMCID: PMC9976740 DOI: 10.1093/ndt/gfac182] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Hypomagnesaemia with secondary hypocal-caemia (HSH) is a rare autosomal recessive disorder caused by pathogenic variants in TRPM6, encoding the channel-kinase transient receptor potential melastatin type 6. Patients have very low serum magnesium (Mg2+) levels and suffer from muscle cramps and seizures. Despite genetic testing, a subgroup of HSH patients remains without a diagnosis. METHODS In this study, two families with an HSH phenotype but negative for TRPM6 pathogenic variants were subjected to whole exome sequencing. Using a complementary combination of biochemical and functional analyses in overexpression systems and patient-derived fibroblasts, the effect of the TRPM7-identified variants on Mg2+ transport was examined. RESULTS For the first time, variants in TRPM7 were identified in two families as a potential cause for hereditary HSH. Patients suffer from seizures and muscle cramps due to magnesium deficiency and episodes of hypocalcaemia. In the first family, a splice site variant caused the incorporation of intron 1 sequences into the TRPM7 messenger RNA and generated a premature stop codon. As a consequence, patient-derived fibroblasts exhibit decreased cell growth. In the second family, a heterozygous missense variant in the pore domain resulted in decreased TRPM7 channel activity. CONCLUSIONS We establish TRPM7 as a prime candidate gene for autosomal dominant hypomagnesaemia and secondary hypocalcaemia. Screening of unresolved patients with hypocalcaemia and secondary hypocalcaemia may further establish TRPM7 pathogenic variants as a novel Mendelian disorder.
Collapse
Affiliation(s)
| | | | | | - Valentina Carotti
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ana Perdomo-Ramirez
- Unidad de Investigación, Renal Tube Group, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - Marguerite Hureaux
- Département de Génétique, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Hôpital Européen Georges Pompidou, Paris, France
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Femke Latta
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pascal Houillier
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, CNRS, Paris, France,Department of Physiology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France,Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Paris, France
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
48
|
Suksridechacin N, Thongon N. Fibroblast growth factor-23 and parathyroid hormone suppress small intestinal magnesium absorption. Physiol Rep 2022; 10:e15247. [PMID: 35385223 PMCID: PMC8985197 DOI: 10.14814/phy2.15247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/22/2022] [Accepted: 03/15/2022] [Indexed: 04/12/2023] Open
Abstract
In the present study, we examined the systemic and direct effects of parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF-23) on duodenal, jejunal, and ileal Mg2+ absorption. The rats were injected with FGF-23 or PTH for 5 h before collecting the duodenum, jejunum, and ileum for Mg2+ transport analysis in Ussing chambers. The duodenum, jejunum, and ileum were directly exposed to FGF-23, PTH, or FGF-23 plus PTH with or without cell signaling inhibitors for 150 min in Ussing chambers prior to performing the Mg2+ transport study. The small intestinal tissues were also subjected to western blot analyses for FGF receptor (FGFR), PTH receptor (PTHR), Klotho, transient receptor potential melastatin 6 (TRPM6), and cyclin as well as the cystathionine β-synthase domain divalent metal cation transport mediator 4 (CNNM4) expression. The small intestine abundantly expressed FGFR and PTHR proteins, whereas, Klotho was not expressed in rat small intestine. Systemic PTH or FGF-23 injection significantly suppressed transcellular Mg2+ transport in the duodenum and jejunum. Direct FGF-23-, PTH-, or FGF-23 plus PTH exposure also suppressed transcellular Mg2+ absorption in the duodenum and jejunum. There was no additional inhibitory effect of PTH and FGF-23 on intestinal Mg2+ absorption. The inhibitory effect of PTH, FGF-23, or FGF-23 plus PTH was abolished by Gö 6850. Systemic PTH- or FGF-23-injection significantly decreased membranous TRPM6 expression, but increased cytosolic CNNM4 expression in the duodenum, jejunum, and ileum. In the present study, we propose a novel magnesiotropic action of PTH and FGF-23 by modulating small intestinal Mg2+ absorption.
Collapse
Affiliation(s)
- Nasisorn Suksridechacin
- Division of PhysiologyDepartment of Biomedical SciencesFaculty of Allied Health SciencesBurapha UniversityChonburiThailand
- Biodiversity Research CentreThailand Institute of Scientific and Technological ResearchPathumthaniThailand
| | - Narongrit Thongon
- Division of PhysiologyDepartment of Biomedical SciencesFaculty of Allied Health SciencesBurapha UniversityChonburiThailand
| |
Collapse
|
49
|
Dent A, Selvaratnam R. Measuring Magnesium – Physiological, Clinical and Analytical Perspectives. Clin Biochem 2022; 105-106:1-15. [DOI: 10.1016/j.clinbiochem.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/23/2022] [Accepted: 04/01/2022] [Indexed: 11/03/2022]
|
50
|
Lo J, Forst AL, Warth R, Zdebik AA. EAST/SeSAME Syndrome and Beyond: The Spectrum of Kir4.1- and Kir5.1-Associated Channelopathies. Front Physiol 2022; 13:852674. [PMID: 35370765 PMCID: PMC8965613 DOI: 10.3389/fphys.2022.852674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
In 2009, two groups independently linked human mutations in the inwardly rectifying K+ channel Kir4.1 (gene name KCNJ10) to a syndrome affecting the central nervous system (CNS), hearing, and renal tubular salt reabsorption. The autosomal recessive syndrome has been named EAST (epilepsy, ataxia, sensorineural deafness, and renal tubulopathy) or SeSAME syndrome (seizures, sensorineural deafness, ataxia, intellectual disability, and electrolyte imbalance), accordingly. Renal dysfunction in EAST/SeSAME patients results in loss of Na+, K+, and Mg2+ with urine, activation of the renin-angiotensin-aldosterone system, and hypokalemic metabolic alkalosis. Kir4.1 is highly expressed in affected organs: the CNS, inner ear, and kidney. In the kidney, it mostly forms heteromeric channels with Kir5.1 (KCNJ16). Biallelic loss-of-function mutations of Kir5.1 can also have disease significance, but the clinical symptoms differ substantially from those of EAST/SeSAME syndrome: although sensorineural hearing loss and hypokalemia are replicated, there is no alkalosis, but rather acidosis of variable severity; in contrast to EAST/SeSAME syndrome, the CNS is unaffected. This review provides a framework for understanding some of these differences and will guide the reader through the growing literature on Kir4.1 and Kir5.1, discussing the complex disease mechanisms and the variable expression of disease symptoms from a molecular and systems physiology perspective. Knowledge of the pathophysiology of these diseases and their multifaceted clinical spectrum is an important prerequisite for making the correct diagnosis and forms the basis for personalized therapies.
Collapse
Affiliation(s)
- Jacky Lo
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Anna-Lena Forst
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Richard Warth
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Anselm A. Zdebik
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- Centre for Nephrology, University College London, London, United Kingdom
| |
Collapse
|