1
|
Tashiro M, Konishi M, Inoue H, Yokoyama U. Selective serotonin reuptake inhibitors (SSRIs) suppress Na +- dependent Mg 2+ efflux in rat ventricular myocytes. J Physiol Sci 2025; 75:100025. [PMID: 40382885 DOI: 10.1016/j.jphyss.2025.100025] [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: 11/18/2024] [Revised: 05/03/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Na+/Mg2+ exchange transport, the Na+ gradient-driven Mg2+ extrusion system, plays a key role in cellular Mg2+ homeostasis. To date, the molecular entity and selective inhibitors of Na+/Mg2+ exchanger have not been fully explored. Intracellular free Mg2+ concentration ([Mg2+]i) was measured in ventricular myocytes acutely isolated from rat hearts. After soaking the cells in high-Mg2+ low-Na+ solution to increase [Mg2+]i, the addition of extracellular Na+ caused a decrease in [Mg2+]i. We analyzed the rate of decrease in [Mg2+]i as Na+/Mg2+ exchange transport activity. The suppression of the rate of decrease in [Mg2+]i caused by sertraline, a selective serotonin reuptake inhibitor (SSRI), was concentration dependent (IC50 8.9 μM) and reversible. Other SSRIs, namely paroxetine and fluvoxamine, were less effective than sertraline. In conclusion, sertraline inhibited Na+/Mg2+ exchange transport more effectively than any previously reported inhibitors of Na+/Mg2+ exchanger. Sertraline could be used as a tool to characterize the functions of Na+/Mg2+ exchanger.
Collapse
Affiliation(s)
- Michiko Tashiro
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan.
| | - Masato Konishi
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Hana Inoue
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan.
| |
Collapse
|
2
|
Franken GAC, Bosman W, Jung HJ, Bos C, Latta F, Knepper M, Hoenderop JGJ, de Baaij JHF. A distal convoluted tubule-specific isoform of murine SLC41A3 extrudes magnesium. Acta Physiol (Oxf) 2025; 241:e70018. [PMID: 39931759 PMCID: PMC11811817 DOI: 10.1111/apha.70018] [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: 08/22/2024] [Revised: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND The distal convoluted tubule (DCT) plays an indispensable role in magnesium (Mg2+) reabsorption in the kidney. Yet, the extrusion mechanism of Mg2+ has not been identified. The solute carrier 41A3 (SLC41A3) has been suggested to be involved in Mg2+ extrusion, but this has never been conclusively demonstrated. METHODS Using available RNA-sequencing data and real-time quantitative PCR, expression of two alternative Slc41a3 transcripts, encoding isoform (Iso) 1 or 2, were assessed in kidney and isolated DCT tubules. HEK293 or HAP1 cells were transfected with plasmids expressing either of the isoforms, followed by 25Mg2+ transport studies. Identification of cis-regulatory elements (CRE) was achieved by combining data from publicly available ATAC sequencing data and luciferase assays. RESULTS Gene expression studies revealed a distinct transcript of Slc41a3 in the DCT with an alternative promoter, leading to a protein with a unique N-terminus; SLC41A3-Iso 2. HEK293 cells overexpressing SLC41A3-Iso 2, but not -Iso 1, exhibited 2.7-fold and 1.6-fold higher 25Mg2+ uptake and extrusion, compared to mock, respectively. The transport was independent of Na+, of the Mg2+ channel TRPM7 or of transporters CNNM3 and -4. We identified a CRE accessible in the DCT, ±2.8kb upstream of the transcript. The presence of the CRE increased the Slc41a3-Iso 2 promoter activity 3.8-fold following luciferase assays, indicating the CRE contains an enhancer function. CONCLUSION In conclusion, we identified two alternative transcripts of Slc41a3 in mouse. Slc41a3-Iso 2 is enriched within the DCT using specific gene regulatory elements. We speculate that specifically in the DCT, SLC41A3-Iso 2 orchestrates Mg2+ extrusion.
Collapse
Affiliation(s)
- Gijs A. C. Franken
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Willem Bosman
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Hyun Jun Jung
- Division of Nephrology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Caro Bos
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Femke Latta
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Mark Knepper
- Epithelial Systems Biology Laboratory, Systems Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Joost G. J. Hoenderop
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Jeroen H. F. de Baaij
- Department of Medical BioSciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
3
|
Jolly JT, Blackburn JS. The PACT Network: PRL, ARL, CNNM, and TRPM Proteins in Magnesium Transport and Disease. Int J Mol Sci 2025; 26:1528. [PMID: 40003994 PMCID: PMC11855589 DOI: 10.3390/ijms26041528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Magnesium, the most abundant divalent metal within the cell, is essential for physiological function and critical in cellular signaling. To maintain cellular homeostasis, intracellular magnesium levels are tightly regulated, as dysregulation is linked to numerous diseases, including cancer, diabetes, cardiovascular disorders, and neurological conditions. Over the past two decades, extensive research on magnesium-regulating proteins has provided valuable insight into their pathogenic and therapeutic potential. This review explores an emerging mechanism of magnesium homeostasis involving proteins in the PRL (phosphatase of regenerating liver), ARL (ADP ribosylation factor-like GTPase family), CNNM (cyclin and cystathionine β-synthase domain magnesium transport mediator), and TRPM (transient receptor potential melastatin) families, collectively termed herein as the PACT network. While each PACT protein has been studied within its individual signaling and disease contexts, their interactions suggest a broader regulatory network with therapeutic potential. This review consolidates the current knowledge on the PACT proteins' structure, function, and interactions and identifies research gaps to encourage future investigation. As the field of magnesium homeostasis continues to advance, understanding PACT protein interactions offers new opportunities for basic research and therapeutic development targeting magnesium-related disorders.
Collapse
Affiliation(s)
- Jeffery T. Jolly
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
- Markey Comprehensive Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jessica S. Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
- Markey Comprehensive Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
4
|
Kröse JL, de Baaij JHF. Magnesium biology. Nephrol Dial Transplant 2024; 39:1965-1975. [PMID: 38871680 PMCID: PMC11648962 DOI: 10.1093/ndt/gfae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Indexed: 06/15/2024] Open
Abstract
Magnesium (Mg2+) is essential for energy metabolism, muscle contraction and neurotransmission. As part of the Mg-ATP complex, it is involved in over 600 enzymatic reactions. Serum Mg2+ levels are tightly regulated between 0.7 and 1.1 mmol/L by interplay of intestinal absorption and renal excretion. In the small intestine, Mg2+ is absorbed paracellularly via claudin-2 and -12. In the colon, transcellular absorption of Mg2+ is facilitated by TRPM6/7 and CNNM4. In the kidney, the proximal tubule reabsorbs only 20% of the filtered Mg2+. The majority of the filtered Mg2+ is reabsorbed in the thick ascending limb, where the lumen-positive transepithelial voltage drives paracellular transport via claudin-16/-19. Fine-tuning of Mg2+ reabsorption is achieved in the distal convoluted tubule (DCT). Here, TRPM6/7 tetramers facilitate apical Mg2+ uptake, which is hormonally regulated by insulin and epidermal growth factor. Basolateral Mg2+ extrusion is Na+ dependent and achieved by CNNM2 and/or SLC41A3. Hypomagnesemia (serum Mg2+ <0.7 mmol/L) develops when intestinal and/or renal Mg2+ (re)absorption is disturbed. Common causes include alcoholism, type 2 diabetes mellitus and the use of pharmacological drugs, such as proton-pump inhibitors, calcineurin inhibitors and thiazide diuretics. Over the last decade, research on rare genetic and acquired Mg2+ disorders have identified Mg2+ channel and transporter activity, DCT length, mitochondrial function and autoimmunity as mechanisms explaining hypomagnesemia. Classically, treatment of hypomagnesemia depended on oral or intravenous Mg2+ supplementation. Recently, prebiotic dietary fibers and sodium-glucose cotransporter 2 inhibitors have been proposed as promising new therapeutic pathways to treat hypomagnesemia.
Collapse
Affiliation(s)
- Jana L Kröse
- Department of Medical BioSciences,
Radboudumc, Nijmegen, The
Netherlands
| | | |
Collapse
|
5
|
Chen YS, Gehring K. New insights into the structure and function of CNNM proteins. FEBS J 2023; 290:5475-5495. [PMID: 37222397 DOI: 10.1111/febs.16872] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/17/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
Magnesium (Mg2+ ) is the most abundant divalent cation in cells and plays key roles in almost all biological processes. CBS-pair domain divalent metal cation transport mediators (CNNMs) are a newly characterized class of Mg2+ transporters present throughout biology. Originally discovered in bacteria, there are four CNNM proteins in humans, which are involved in divalent cation transport, genetic diseases, and cancer. Eukaryotic CNNMs are composed of four domains: an extracellular domain, a transmembrane domain, a cystathionine-β-synthase (CBS)-pair domain, and a cyclic nucleotide-binding homology domain. The transmembrane and CBS-pair core are the defining features of CNNM proteins with over 20 000 protein sequences known from over 8000 species. Here, we review the structural and functional studies of eukaryotic and prokaryotic CNNMs that underlie our understanding of their regulation and mechanism of ion transport. Recent structures of prokaryotic CNNMs confirm the transmembrane domain mediates ion transport with the CBS-pair domain likely playing a regulatory role through binding divalent cations. Studies of mammalian CNNMs have identified new binding partners. These advances are driving progress in understanding this deeply conserved and widespread family of ion transporters.
Collapse
Affiliation(s)
- Yu Seby Chen
- Department of Biochemistry & Molecular Biology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Kalle Gehring
- Department of Biochemistry & Centre de Recherche en Biologie Structurale, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Hardy S, Zolotarov Y, Coleman J, Roitman S, Khursheed H, Aubry I, Uetani N, Tremblay M. PRL-1/2 phosphatases control TRPM7 magnesium-dependent function to regulate cellular bioenergetics. Proc Natl Acad Sci U S A 2023; 120:e2221083120. [PMID: 36972446 PMCID: PMC10083557 DOI: 10.1073/pnas.2221083120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Phosphatases of regenerating liver (PRL-1, PRL-2, PRL-3; also known as PTP4A1, PTP4A2, PTP4A3, respectively) control intracellular magnesium levels by interacting with the CNNM magnesium transport regulators. Still, the exact mechanism governing magnesium transport by this protein complex is not well understood. Herein, we have developed a genetically encoded intracellular magnesium-specific reporter and demonstrate that the CNNM family inhibits the function of the TRPM7 magnesium channel. We show that the small GTPase ARL15 increases CNNM3/TRPM7 protein complex formation to reduce TRPM7 activity. Conversely, PRL-2 overexpression counteracts ARL15 binding to CNNM3 and enhances the function of TRPM7 by preventing the interaction between CNNM3 and TRPM7. Moreover, while TRPM7-induced cell signaling is promoted by PRL-1/2, it is reduced when CNNM3 is overexpressed. Lowering cellular magnesium levels reduces the interaction of CNNM3 with TRPM7 in a PRL-dependent manner, whereby knockdown of PRL-1/2 restores the protein complex formation. Cotargeting of TRPM7 and PRL-1/2 alters mitochondrial function and sensitizes cells to metabolic stress induced by magnesium depletion. These findings reveal the dynamic regulation of TRPM7 function in response to PRL-1/2 levels, to coordinate magnesium transport and reprogram cellular metabolism.
Collapse
Affiliation(s)
- Serge Hardy
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Yevgen Zolotarov
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Jacob Coleman
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Simon Roitman
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Hira Khursheed
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Isabelle Aubry
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Noriko Uetani
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Michel L. Tremblay
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| |
Collapse
|
7
|
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
|
8
|
Funato Y, Hashizume O, Miki H. Phosphatase-independent role of phosphatase of regenerating liver in cancer progression. Cancer Sci 2022; 114:25-33. [PMID: 36285487 PMCID: PMC9807511 DOI: 10.1111/cas.15625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 01/07/2023] Open
Abstract
Phosphatase of regenerating liver (PRL) is a family of protein tyrosine phosphatases (PTPs) that are anchored to the plasma membrane by prenylation. They are frequently overexpressed in various types of malignant cancers and their roles in cancer progression have received considerable attention. Mutational analyses of PRLs have shown that their intrinsic phosphatase activity is dispensable for tumor formation induced by PRL overexpression in a lung metastasis model using melanoma cells. Instead, PRLs directly bind to cyclin M (CNNM) Mg2+ exporters in the plasma membrane and potently inhibit their Mg2+ export activity, resulting in an increase in intracellular Mg2+ levels. Experiments using mammalian culture cells, mice, and C. elegans have collectively revealed that dysregulation of Mg2+ levels severely affects ATP and reactive oxygen species (ROS) levels as well as the function of Ca2+ -permeable channels. Moreover, PRL overexpression altered the optimal pH for cell proliferation from normal 7.5 to acidic 6.5, which is typically observed in malignant tumors. Here, we review the phosphatase-independent biological functions of PRLs, focusing on their interactions with CNNM Mg2+ exporters in cancer progression.
Collapse
Affiliation(s)
- Yosuke Funato
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Osamu Hashizume
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Hiroaki Miki
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversityOsakaJapan,Center for Infectious Disease Education and Research (CiDER)Osaka UniversityOsakaJapan
| |
Collapse
|
9
|
Inoue S, Hayashi M, Huang S, Yokosho K, Gotoh E, Ikematsu S, Okumura M, Suzuki T, Kamura T, Kinoshita T, Ma JF. A tonoplast-localized magnesium transporter is crucial for stomatal opening in Arabidopsis under high Mg 2+ conditions. THE NEW PHYTOLOGIST 2022; 236:864-877. [PMID: 35976788 PMCID: PMC9804957 DOI: 10.1111/nph.18410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Plant stomata play an important role in CO2 uptake for photosynthesis and transpiration, but the mechanisms underlying stomatal opening and closing under changing environmental conditions are still not completely understood. Through large-scale genetic screening, we isolated an Arabidopsis mutant (closed stomata2 (cst2)) that is defective in stomatal opening. We cloned the causal gene (MGR1/CST2) and functionally characterized this gene. The mutant phenotype was caused by a mutation in a gene encoding an unknown protein with similarities to the human magnesium (Mg2+ ) efflux transporter ACDP/CNNM. MGR1/CST2 was localized to the tonoplast and showed transport activity for Mg2+ . This protein was constitutively and highly expressed in guard cells. Knockout of this gene resulted in stomatal closing, decreased photosynthesis and growth retardation, especially under high Mg2+ conditions, while overexpression of this gene increased stomatal opening and tolerance to high Mg2+ concentrations. Furthermore, guard cell-specific expression of MGR1/CST2 in the mutant partially restored its stomatal opening. Our results indicate that MGR1/CST2 expression in the leaf guard cells plays an important role in maintaining cytosolic Mg2+ concentrations through sequestering Mg2+ into vacuoles, which is required for stomatal opening, especially under high Mg2+ conditions.
Collapse
Affiliation(s)
- Shin‐ichiro Inoue
- Division of Biological Science, Graduate School of ScienceNagoya UniversityFuro‐cho, Chikusa‐kuNagoyaAichi464‐8602Japan
| | - Maki Hayashi
- Division of Biological Science, Graduate School of ScienceNagoya UniversityFuro‐cho, Chikusa‐kuNagoyaAichi464‐8602Japan
| | - Sheng Huang
- Institute of Plant Science and ResourcesOkayama UniversityChuo 2‐20‐1Kurashiki710‐0046Japan
| | - Kengo Yokosho
- Institute of Plant Science and ResourcesOkayama UniversityChuo 2‐20‐1Kurashiki710‐0046Japan
| | - Eiji Gotoh
- Department of Forest Environmental Sciences, Faculty of AgricultureKyushu University744 MotookaFukuoka819‐0395Japan
| | - Shuka Ikematsu
- Institute of Transformative Bio‐Molecules (WPI‐ITbM)Nagoya UniversityFuro‐cho, ChikusaNagoya464‐8602Japan
| | - Masaki Okumura
- Division of Biological Science, Graduate School of ScienceNagoya UniversityFuro‐cho, Chikusa‐kuNagoyaAichi464‐8602Japan
| | - Takamasa Suzuki
- Department of Biological Chemistry, College of Bioscience and BiotechnologyChubu UniversityKasugai‐shiAichi487‐8501Japan
| | - Takumi Kamura
- Division of Biological Science, Graduate School of ScienceNagoya UniversityFuro‐cho, Chikusa‐kuNagoyaAichi464‐8602Japan
| | - Toshinori Kinoshita
- Division of Biological Science, Graduate School of ScienceNagoya UniversityFuro‐cho, Chikusa‐kuNagoyaAichi464‐8602Japan
- Institute of Transformative Bio‐Molecules (WPI‐ITbM)Nagoya UniversityFuro‐cho, ChikusaNagoya464‐8602Japan
| | - Jian Feng Ma
- Institute of Plant Science and ResourcesOkayama UniversityChuo 2‐20‐1Kurashiki710‐0046Japan
| |
Collapse
|
10
|
Gyimesi G, Hediger MA. Systematic in silico discovery of novel solute carrier-like proteins from proteomes. PLoS One 2022; 17:e0271062. [PMID: 35901096 PMCID: PMC9333335 DOI: 10.1371/journal.pone.0271062] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/22/2022] [Indexed: 12/26/2022] Open
Abstract
Solute carrier (SLC) proteins represent the largest superfamily of transmembrane transporters. While many of them play key biological roles, their systematic analysis has been hampered by their functional and structural heterogeneity. Based on available nomenclature systems, we hypothesized that many as yet unidentified SLC transporters exist in the human genome, which await further systematic analysis. Here, we present criteria for defining "SLC-likeness" to curate a set of "SLC-like" protein families from the Transporter Classification Database (TCDB) and Protein families (Pfam) databases. Computational sequence similarity searches surprisingly identified ~120 more proteins in human with potential SLC-like properties compared to previous annotations. Interestingly, several of these have documented transport activity in the scientific literature. To complete the overview of the "SLC-ome", we present an algorithm to classify SLC-like proteins into protein families, investigating their known functions and evolutionary relationships to similar proteins from 6 other clinically relevant experimental organisms, and pinpoint structural orphans. We envision that our work will serve as a stepping stone for future studies of the biological function and the identification of the natural substrates of the many under-explored SLC transporters, as well as for the development of new therapeutic applications, including strategies for personalized medicine and drug delivery.
Collapse
Affiliation(s)
- Gergely Gyimesi
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department for BioMedical Research, Inselspital, University of Bern, Bern, Switzerland
- * E-mail: (GG); (MAH)
| | - Matthias A. Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department for BioMedical Research, Inselspital, University of Bern, Bern, Switzerland
- * E-mail: (GG); (MAH)
| |
Collapse
|
11
|
Tseng MH, Yang SS, Sung CC, Ding JJ, Hsu YJ, Chu SM, Lin SH. Novel CNNM2 Mutation Responsible for Autosomal-Dominant Hypomagnesemia With Seizure. Front Genet 2022; 13:875013. [PMID: 35846113 PMCID: PMC9277586 DOI: 10.3389/fgene.2022.875013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
CNNM2 is primarily expressed in the brain and distal convoluted tubule (DCT) of the kidney. Mutations in CNNM2 have been reported to cause hypomagnesemia, seizure, and intellectual disability (HSMR) syndrome. However, the clinical and functional effect of CNNM2 mutations remains incompletely understood. We report our clinical encounter with a 1-year-old infant with HSMR features. Mutation screening for this trio family was performed using next-generation sequencing (NGS)-based whole exome sequencing (WES) with the identified mutation verified by Sanger sequencing. We identified a de novo heterozygous mutation c.G1439T (R480L) in the essential cystathionine β-synthase (CBS) domain of CNNM2 encoding CNNM2 (cyclin M2) without any other gene mutations related to hypomagnesemia. The amino acid involved in this missense mutation was conserved in different species. It was also found to be pathogenic based on the different software prediction models and ACGME criteria. In vitro studies revealed a higher expression of the CNNM2-R480L mutant protein compared to that of the wild-type CNNM2. Like the CNNM2-wild type, proper localization of CNNM2-R480L was shown on immunocytochemistry images. The Mg2+ efflux assay in murine DCT (mDCT) cells revealed a significant increase in intracellular Mg2+ green in CNNM2-R480L compared to that in CNNM2-WT. By using a simulation model, we illustrate that the R480L mutation impaired the interaction between CNNM2 and ATP-Mg2+. We propose that this novel R480L mutation in the CNNM2 gene led to impaired binding between Mg2+-ATP and CNNM2 and diminished Mg2+ efflux, manifesting clinically as refractory hypomagnesemia.
Collapse
Affiliation(s)
- Min-Hua Tseng
- Division of Nephrology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.,Department of Pediatrics, Xiamen Chang Gung Hospital, Ximen, China
| | - Sung-Sen Yang
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Chien Sung
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jhao-Jhuang Ding
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Chu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
12
|
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
|
13
|
Franken GAC, Huynen MA, Martínez-Cruz LA, Bindels RJM, de Baaij JHF. Structural and functional comparison of magnesium transporters throughout evolution. Cell Mol Life Sci 2022; 79:418. [PMID: 35819535 PMCID: PMC9276622 DOI: 10.1007/s00018-022-04442-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/22/2022] [Accepted: 06/21/2022] [Indexed: 12/16/2022]
Abstract
Magnesium (Mg2+) is the most prevalent divalent intracellular cation. As co-factor in many enzymatic reactions, Mg2+ is essential for protein synthesis, energy production, and DNA stability. Disturbances in intracellular Mg2+ concentrations, therefore, unequivocally result in delayed cell growth and metabolic defects. To maintain physiological Mg2+ levels, all organisms rely on balanced Mg2+ influx and efflux via Mg2+ channels and transporters. This review compares the structure and the function of prokaryotic Mg2+ transporters and their eukaryotic counterparts. In prokaryotes, cellular Mg2+ homeostasis is orchestrated via the CorA, MgtA/B, MgtE, and CorB/C Mg2+ transporters. For CorA, MgtE, and CorB/C, the motifs that form the selectivity pore are conserved during evolution. These findings suggest that CNNM proteins, the vertebrate orthologues of CorB/C, also have Mg2+ transport capacity. Whereas CorA and CorB/C proteins share the gross quaternary structure and functional properties with their respective orthologues, the MgtE channel only shares the selectivity pore with SLC41 Na+/Mg2+ transporters. In eukaryotes, TRPM6 and TRPM7 Mg2+ channels provide an additional Mg2+ transport mechanism, consisting of a fusion of channel with a kinase. The unique features these TRP channels allow the integration of hormonal, cellular, and transcriptional regulatory pathways that determine their Mg2+ transport capacity. Our review demonstrates that understanding the structure and function of prokaryotic magnesiotropic proteins aids in our basic understanding of Mg2+ transport.
Collapse
Affiliation(s)
- G A C Franken
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - M A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L A Martínez-Cruz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park, Derio, 48160, Bizkaia, Spain
| | - R J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - J H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Cavarocchi E, Whitfield M, Saez F, Touré A. Sperm Ion Transporters and Channels in Human Asthenozoospermia: Genetic Etiology, Lessons from Animal Models, and Clinical Perspectives. Int J Mol Sci 2022; 23:ijms23073926. [PMID: 35409285 PMCID: PMC8999829 DOI: 10.3390/ijms23073926] [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: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022] Open
Abstract
In mammals, sperm fertilization potential relies on efficient progression within the female genital tract to reach and fertilize the oocyte. This fundamental property is supported by the flagellum, an evolutionarily conserved organelle that provides the mechanical force for sperm propulsion and motility. Importantly several functional maturation events that occur during the journey of the sperm cells through the genital tracts are necessary for the activation of flagellar beating and the acquisition of fertilization potential. Ion transporters and channels located at the surface of the sperm cells have been demonstrated to be involved in these processes, in particular, through the activation of downstream signaling pathways and the promotion of novel biochemical and electrophysiological properties in the sperm cells. We performed a systematic literature review to describe the currently known genetic alterations in humans that affect sperm ion transporters and channels and result in asthenozoospermia, a pathophysiological condition defined by reduced or absent sperm motility and observed in nearly 80% of infertile men. We also present the physiological relevance and functional mechanisms of additional ion channels identified in the mouse. Finally, considering the state-of-the art, we discuss future perspectives in terms of therapeutics of asthenozoospermia and male contraception.
Collapse
Affiliation(s)
- Emma Cavarocchi
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
| | - Marjorie Whitfield
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
| | - Fabrice Saez
- UMR GReD Institute (Génétique Reproduction & Développement) CNRS 6293, INSERM U1103, Team «Mécanismes de L’Infertilité Mâle Post-Testiculaire», Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Correspondence: (F.S.); (A.T.)
| | - Aminata Touré
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
- Correspondence: (F.S.); (A.T.)
| |
Collapse
|
15
|
Xu X, Hou S, Sun W, Zhu J, Yuan J, Cui Z, Wu D, Tang J. Rare hypomagnesemia, seizures, and mental retardation in a 4-month-old patient caused by novel CNNM2 mutation Tyr189Cys: Genetic analysis and review. Mol Genet Genomic Med 2022; 10:e1898. [PMID: 35170241 PMCID: PMC9000947 DOI: 10.1002/mgg3.1898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/24/2021] [Accepted: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
Background Hypomagnesemia, seizures, and mental retardation (HSMR) syndrome is a rare genetic disease. Presently, only 24 cases have been reported and the clinical features of the disease are yet to be fully described, thereby making diagnosis challenging. Methods Trio‐whole‐exome sequencing was used for the patient and her parents, and the structure of the variant protein was analyzed by molecular dynamics. Finally, the characteristics of HSMR were summarized by reviewing the previous literature. Results The main disease manifestations in the patient were seizures, liver function damage, hypomagnesemia, atrial septal defect, and sinus arrhythmia. A novel mutation in CNNM2 (c.566A>G/p.Tyr189Cys) was identified by genetic detection. The parents were wild type, and the mutation was rated as pathogenic by American College of Medical Genetics and Genomics guidelines. Ab initio modeling and molecular dynamics simulation show that the mutation destroys the surrounding hydrogen bonds, which may reduce the local stability of the protein structure. In the previous literature, only 24 children with HSMR have been reported, mainly manifested as hypomagnesemia, mental retardation, seizures, and language and motor impairment. Conclusion We have reported the second case of HSMR in the Chinese population, which further expands the phenotypic spectrum of congenital heart disease and the variation spectrum of CNNM2.
Collapse
Affiliation(s)
- Xiaoyan Xu
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shu Hou
- Department of Pediatrics, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weiwei Sun
- Beijing Chigene Translational Medical Research Center Co. Ltd, Beijing, China
| | - Jing Zhu
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinjing Yuan
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenzhen Cui
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De Wu
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiulai Tang
- Department of Pediatrics, Neurological Rehabilitation Center, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Reyes JV, Medina PMB. Renal calcium and magnesium handling in Gitelman syndrome. Am J Transl Res 2022; 14:1-19. [PMID: 35173827 PMCID: PMC8829599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/24/2021] [Indexed: 06/14/2023]
Abstract
Gitelman syndrome (GS) is an autosomal recessive salt-losing tubulopathy caused by biallelic inactivating mutations in the SLC12A3 gene. This gene encodes the thiazide-sensitive sodium-chloride cotransporter (NCC) which is exclusively expressed in the distal convoluted tubules (DCT). GS patients classically present with hypokalemic metabolic alkalosis with hypocalciuria and hypomagnesemia. While hypokalemia and metabolic alkalosis are easily explained by effects of the genotypic defect in GS, the mechanisms by which hypomagnesemia and hypocalciuria develop in GS are poorly understood. In this review, we aim to achieve three major objectives. First, present a concise discussion about current understanding on physiologic calcium and magnesium handling in the DCT. Second, integrate expression data from studies on calciotropic and magnesiotropic proteins relevant to the GS disease state. Lastly, provide insights into the possible mechanisms of calcium-magnesium crosstalk relating to the co-occurrence of hypocalciuria and hypomagnesemia in GS models. Our analyses highlight specific areas of study that are valuable in elucidating possible molecular pathways of hypocalciuria and hypomagnesemia in GS.
Collapse
Affiliation(s)
- Jeremiah V Reyes
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila Ermita, Manila 1000, Philippines
| | - Paul Mark B Medina
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila Ermita, Manila 1000, Philippines
| |
Collapse
|
17
|
Bai Z, Feng J, Franken GAC, Al’Saadi N, Cai N, Yu AS, Lou L, Komiya Y, Hoenderop JGJ, de Baaij JHF, Yue L, Runnels LW. CNNM proteins selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. PLoS Biol 2021; 19:e3001496. [PMID: 34928937 PMCID: PMC8726484 DOI: 10.1371/journal.pbio.3001496] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 01/04/2022] [Accepted: 11/26/2021] [Indexed: 01/04/2023] Open
Abstract
Magnesium is essential for cellular life, but how it is homeostatically controlled still remains poorly understood. Here, we report that members of CNNM family, which have been controversially implicated in both cellular Mg2+ influx and efflux, selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. Coexpression of CNNMs with the channel markedly increased uptake of divalent cations, which is prevented by an inactivating mutation to the channel’s pore. Knockout (KO) of TRPM7 in cells or application of the TRPM7 channel inhibitor NS8593 also interfered with CNNM-stimulated divalent cation uptake. Conversely, KO of CNNM3 and CNNM4 in HEK-293 cells significantly reduced TRPM7-mediated divalent cation entry, without affecting TRPM7 protein expression or its cell surface levels. Furthermore, we found that cellular overexpression of phosphatases of regenerating liver (PRLs), known CNNMs binding partners, stimulated TRPM7-dependent divalent cation entry and that CNNMs were required for this activity. Whole-cell electrophysiological recordings demonstrated that deletion of CNNM3 and CNNM4 from HEK-293 cells interfered with heterologously expressed and native TRPM7 channel function. We conclude that CNNMs employ the TRPM7 channel to mediate divalent cation influx and that CNNMs also possess separate TRPM7-independent Mg2+ efflux activities that contribute to CNNMs’ control of cellular Mg2+ homeostasis. Magnesium is essential for cellular life, but how is it homeostatically controlled? This study shows that proteins of the CNNM family bind to the TRPM7 channel to stimulate divalent cation entry into cells, independent of their function in regulating magnesium ion efflux.
Collapse
Affiliation(s)
- Zhiyong Bai
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Jianlin Feng
- UCONN Health Center, Farmington, New Mexico, United States of America
| | | | - Namariq Al’Saadi
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
- University of Misan, Amarah, Iraq
| | - Na Cai
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Albert S. Yu
- UCONN Health Center, Farmington, New Mexico, United States of America
| | - Liping Lou
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Yuko Komiya
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | | | | | - Lixia Yue
- UCONN Health Center, Farmington, New Mexico, United States of America
| | - Loren W. Runnels
- Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
18
|
Kollewe A, Chubanov V, Tseung FT, Correia L, Schmidt E, Rössig A, Zierler S, Haupt A, Müller CS, Bildl W, Schulte U, Nicke A, Fakler B, Gudermann T. The molecular appearance of native TRPM7 channel complexes identified by high-resolution proteomics. eLife 2021; 10:68544. [PMID: 34766907 PMCID: PMC8616561 DOI: 10.7554/elife.68544] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed membrane protein consisting of ion channel and protein kinase domains. TRPM7 plays a fundamental role in the cellular uptake of divalent cations such as Zn2+, Mg2+, and Ca2+, and thus shapes cellular excitability, plasticity, and metabolic activity. The molecular appearance and operation of TRPM7 channels in native tissues have remained unresolved. Here, we investigated the subunit composition of endogenous TRPM7 channels in rodent brain by multi-epitope affinity purification and high-resolution quantitative mass spectrometry (MS) analysis. We found that native TRPM7 channels are high-molecular-weight multi-protein complexes that contain the putative metal transporter proteins CNNM1-4 and a small G-protein ADP-ribosylation factor-like protein 15 (ARL15). Heterologous reconstitution experiments confirmed the formation of TRPM7/CNNM/ARL15 ternary complexes and indicated that complex formation effectively and specifically impacts TRPM7 activity. These results open up new avenues towards a mechanistic understanding of the cellular regulation and function of TRPM7 channels.
Collapse
Affiliation(s)
- Astrid Kollewe
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Fong Tsuen Tseung
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Leonor Correia
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Eva Schmidt
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Anna Rössig
- 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
| | - Alexander Haupt
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Catrin Swantje Müller
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Annette Nicke
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Bernd Fakler
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,German Center for Lung Research, Munich, Germany
| |
Collapse
|
19
|
Claverie-Martin F, Perdomo-Ramirez A, Garcia-Nieto V. Hereditary kidney diseases associated with hypomagnesemia. Kidney Res Clin Pract 2021; 40:512-526. [PMID: 34784661 PMCID: PMC8685365 DOI: 10.23876/j.krcp.21.112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/20/2021] [Indexed: 11/04/2022] Open
Abstract
In the kidney, a set of proteins expressed in the epithelial cells of the thick ascending loop of Henle and the distal convoluted tubule directly or indirectly play important roles in the regulation of serum magnesium levels. Magnesium reabsorption in the thick ascending loop of Henle occurs through a passive paracellular pathway, while in the distal convoluted tubule, the final magnesium concentration is established through an active transcellular pathway. The players involved in magnesium reabsorption include proteins with diverse functions including tight junction proteins, cation and anion channels, sodium chloride cotransporter, calcium-sensing receptor, epidermal growth factor, cyclin M2, sodium potassium adenosine triphosphatase subunits, transcription factors, a serine protease, and proteins involved in mitochondrial function. Mutations in the genes that encode these proteins impair their function and cause different rare diseases associated with hypomagnesemia, which may lead to muscle cramps, fatigue, epileptic seizures, intellectual disability, cardiac arrhythmias, and chronic kidney disease. The purpose of this review is to describe the clinical and genetic characteristics of these hereditary kidney diseases and the current research findings on the pathophysiological basis of these diseases.
Collapse
Affiliation(s)
- Felix Claverie-Martin
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Ana Perdomo-Ramirez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Victor Garcia-Nieto
- Unidad de Nefrología Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| |
Collapse
|
20
|
Tseng MH, Konrad M, Ding JJ, Lin SH. Clinical and Genetic Approach to Renal Hypomagnesemia. Biomed J 2021; 45:74-87. [PMID: 34767995 PMCID: PMC9133307 DOI: 10.1016/j.bj.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Magnesium (Mg2+) is an important intracellular cation and essential to maintain cell function including cell proliferation, immunity, cellular energy metabolism, protein and nucleic acid synthesis, and regulation of ion channels. Consequences of hypomagnesemia affecting multiple organs can be in overt or subtle presentations. Besides detailed history and complete physical examination, the assessment of urinary Mg2+ excretion is help to differentiate renal from extra-renal (gastrointestinal, tissue sequestration, and shifting) causes of hypomagnesemia. Renal hypomagnesemia can be caused by an increased glomerular filtration and impaired reabsorption in proximal tubular cells, thick ascending limb of the loop of Henle or distal convoluted tubules. A combination of renal Mg2+ wasting, familial history, age of onset, associated features, and exclusion of acquired etiologies point to inherited forms of renal hypomagnesemia. Based on clinical phenotypes, its definite genetic diagnosis can be simply grouped into specific, uncertain, and unknown gene mutations with a priority of genetic approach methods. An unequivocal molecular diagnosis could allow for prediction of clinical outcome, providing genetic counseling, avoiding unnecessary studies or interventions, and possibly uncovering the pathogenic mechanism. Given numerous identified genes responsible for Mg2+ transport in renal hypomagnesemia over the past two decades, several potential and specific molecular and cellular therapeutic strategies to correct hypomagnesemia are promising.
Collapse
Affiliation(s)
- Min-Hua Tseng
- Division of Nephrology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, Xiamen Chang Gung Hospital, China
| | - Martin Konrad
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Jhao-Jhuang Ding
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
21
|
Ellison DH, Maeoka Y, McCormick JA. Molecular Mechanisms of Renal Magnesium Reabsorption. J Am Soc Nephrol 2021; 32:2125-2136. [PMID: 34045316 PMCID: PMC8729834 DOI: 10.1681/asn.2021010042] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 02/04/2023] Open
Abstract
Magnesium is an essential cofactor in many cellular processes, and aberrations in magnesium homeostasis can have life-threatening consequences. The kidney plays a central role in maintaining serum magnesium within a narrow range (0.70-1.10 mmol/L). Along the proximal tubule and thick ascending limb, magnesium reabsorption occurs via paracellular pathways. Members of the claudin family form the magnesium pores in these segments, and also regulate magnesium reabsorption by adjusting the transepithelial voltage that drives it. Along the distal convoluted tubule transcellular reabsorption via heteromeric TRPM6/7 channels predominates, although paracellular reabsorption may also occur. In this segment, the NaCl cotransporter plays a critical role in determining transcellular magnesium reabsorption. Although the general machinery involved in renal magnesium reabsorption has been identified by studying genetic forms of magnesium imbalance, the mechanisms regulating it are poorly understood. This review discusses pathways of renal magnesium reabsorption by different segments of the nephron, emphasizing newer findings that provide insight into regulatory process, and outlining critical unanswered questions.
Collapse
Affiliation(s)
- David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon,Veterans Affairs Portland Healthcare System, Portland, Oregon
| | - Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A. McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
22
|
Yin M, Matsuoka R, Xi Y, Wang X. Comparison of Egg Yolk and Soybean Phospholipids on Hepatic Fatty Acid Profile and Liver Protection in Rats Fed a High-Fructose Diet. Foods 2021; 10:1569. [PMID: 34359438 PMCID: PMC8307941 DOI: 10.3390/foods10071569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/30/2022] Open
Abstract
Perturbed lipid metabolism leads to ectopic lipid accumulation in tissues, such as the liver, thereby causing nonalcoholic fatty liver disease (NAFLD) and negatively influencing circulating lipid profile-inducing dyslipidemia. Phospholipids (PLs) with special biological activity are used to treat chronic diseases such as cardiovascular and cerebrovascular disease. PLs derived from egg yolk and soya bean have significant antioxidant and lipid-lowering abilities. This study examined the therapeutic effects of them on hyperlipidemia using a high-fructose-fed rat model; lipid metabolism and anti-inflammatory effects were also analyzed. The results showed that both egg yolk and soya bean phospholipids (EPLs and SPLs) reduced liver weight, hepatic TG, and MDA content as well as serum ALT, AST, TBA, and CRP levels (p < 0.05). The PLs also showed hypolipidemic and anti-inflammatory effects. EPLs and SPLs could inhibit the accumulation of hepatic fatty acids C18:1N9C, C18:0, and C22:6NS of rats fed a high-fat-and-sucrose diet. The intake of EPLs could significantly increase acetylcholine content in the blood and brain tissue. Histological examination showed that PLs intake could ameliorate the damage to liver tissue. This study suggested that EPLs and SPLs had a certain capacity of hypolipidemic and liver protection, and the therapeutic benefits of EPLs tended to be more effective than that of soybean phospholipids.
Collapse
Affiliation(s)
- Mingyu Yin
- College of Food Science and Technology, Shanghai Ocean University, No. 999, Huchenghuan Rd, Nanhui New City, Pudong New District, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai 201306, China
| | - Ryosuke Matsuoka
- College of Food Science and Technology, Shanghai Ocean University, No. 999, Huchenghuan Rd, Nanhui New City, Pudong New District, Shanghai 201306, China
| | - Yinci Xi
- College of Food Science and Technology, Shanghai Ocean University, No. 999, Huchenghuan Rd, Nanhui New City, Pudong New District, Shanghai 201306, China
| | - Xichang Wang
- College of Food Science and Technology, Shanghai Ocean University, No. 999, Huchenghuan Rd, Nanhui New City, Pudong New District, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai 201306, China
| |
Collapse
|
23
|
Abstract
CNNM/CorB proteins are a broadly conserved family of integral membrane proteins with close to 90,000 protein sequences known. They are associated with Mg2+ transport but it is not known if they mediate transport themselves or regulate other transporters. Here, we determine the crystal structure of an archaeal CorB protein in two conformations (apo and Mg2+-ATP bound). The transmembrane DUF21 domain exists in an inward-facing conformation with a Mg2+ ion coordinated by a conserved π-helix. In the absence of Mg2+-ATP, the CBS-pair domain adopts an elongated dimeric configuration with previously unobserved domain-domain contacts. Hydrogen-deuterium exchange mass spectrometry, analytical ultracentrifugation, and molecular dynamics experiments support a role of the structural rearrangements in mediating Mg2+-ATP sensing. Lastly, we use an in vitro, liposome-based assay to demonstrate direct Mg2+ transport by CorB proteins. These structural and functional insights provide a framework for understanding function of CNNMs in Mg2+ transport and associated diseases.
Collapse
|
24
|
ARL15 modulates magnesium homeostasis through N-glycosylation of CNNMs. Cell Mol Life Sci 2021; 78:5427-5445. [PMID: 34089346 PMCID: PMC8257531 DOI: 10.1007/s00018-021-03832-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cyclin M (CNNM1-4) proteins maintain cellular and body magnesium (Mg2+) homeostasis. Using various biochemical approaches, we have identified members of the CNNM family as direct interacting partners of ADP-ribosylation factor-like GTPase 15 (ARL15), a small GTP-binding protein. ARL15 interacts with CNNMs at their carboxyl-terminal conserved cystathionine-β-synthase (CBS) domains. In silico modeling of the interaction between CNNM2 and ARL15 supports that the small GTPase specifically binds the CBS1 and CNBH domains. Immunocytochemical experiments demonstrate that CNNM2 and ARL15 co-localize in the kidney, with both proteins showing subcellular localization in the endoplasmic reticulum, Golgi apparatus and the plasma membrane. Most importantly, we found that ARL15 is required for forming complex N-glycosylation of CNNMs. Overexpression of ARL15 promotes complex N-glycosylation of CNNM3. Mg2+ uptake experiments with a stable isotope demonstrate that there is a significant increase of 25Mg2+ uptake upon knockdown of ARL15 in multiple kidney cancer cell lines. Altogether, our results establish ARL15 as a novel negative regulator of Mg2+ transport by promoting the complex N-glycosylation of CNNMs.
Collapse
|
25
|
Abstract
Magnesium (Mg2+) plays an essential role in many biological processes. Mg2+ deficiency is therefore associated with a wide range of clinical effects including muscle cramps, fatigue, seizures and arrhythmias. To maintain sufficient Mg2+ levels, (re)absorption of Mg2+ in the intestine and kidney is tightly regulated. Genetic defects that disturb Mg2+ uptake pathways, as well as drugs interfering with Mg2+ (re)absorption cause hypomagnesemia. The aim of this review is to provide an overview of the molecular mechanisms underlying genetic and drug-induced Mg2+ deficiencies. This leads to the identification of four main mechanisms that are affected by hypomagnesemia-causing mutations or drugs: luminal transient receptor potential melastatin type 6/7-mediated Mg2+ uptake, paracellular Mg2+ reabsorption in the thick ascending limb of Henle's loop, structural integrity of the distal convoluted tubule and Na+-dependent Mg2+ extrusion driven by the Na+/K+-ATPase. Our analysis demonstrates that genetic and drug-induced causes of hypomagnesemia share common molecular mechanisms. Targeting these shared pathways can lead to novel treatment options for patients with hypomagnesemia.
Collapse
|
26
|
Huang Y, Mu K, Teng X, Zhao Y, Funato Y, Miki H, Zhu W, Xu Z, Hattori M. Identification and mechanistic analysis of an inhibitor of the CorC Mg 2+ transporter. iScience 2021; 24:102370. [PMID: 33912817 PMCID: PMC8066426 DOI: 10.1016/j.isci.2021.102370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 11/17/2022] Open
Abstract
The CorC/CNNM family of Na+-dependent Mg2+ transporters is ubiquitously conserved from bacteria to humans. CorC, the bacterial CorC/CNNM family of proteins, is involved in resistance to antibiotic exposure and in the survival of pathogenic microorganisms in their host environment. The CorC/CNNM family proteins possess a cytoplasmic region containing the regulatory ATP-binding site. CorC and CNNM have attracted interest as therapeutic targets, whereas inhibitors targeting the ATP-binding site have not been identified. Here, we performed a virtual screening of CorC by targeting its ATP-binding site, identified a compound named IGN95a with inhibitory effects on ATP binding and Mg2+ export, and determined the cytoplasmic domain structure in complex with IGN95a. Furthermore, a chemical cross-linking experiment indicated that with ATP bound to the cytoplasmic domain, the conformational equilibrium of CorC was shifted more toward the inward-facing state of the transmembrane domain. In contrast, IGN95a did not induce such a shift.
Collapse
Affiliation(s)
- Yichen Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Kaijie Mu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong New Area, Shanghai, 201203, China
| | - Xinyu Teng
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Yosuke Funato
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroaki Miki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong New Area, Shanghai, 201203, China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong New Area, Shanghai, 201203, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| |
Collapse
|
27
|
Cyclin M2 (CNNM2) knockout mice show mild hypomagnesaemia and developmental defects. Sci Rep 2021; 11:8217. [PMID: 33859252 PMCID: PMC8050252 DOI: 10.1038/s41598-021-87548-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/26/2021] [Indexed: 02/02/2023] Open
Abstract
Patients with mutations in Cyclin M2 (CNNM2) suffer from hypomagnesaemia, seizures, and intellectual disability. Although the molecular function of CNNM2 is under debate, the protein is considered essential for renal Mg2+ reabsorption. Here, we used a Cnnm2 knock out mouse model, generated by CRISPR/Cas9 technology, to assess the role of CNNM2 in Mg2+ homeostasis. Breeding Cnnm2+/- mice resulted in a Mendelian distribution at embryonic day 18. Nevertheless, only four Cnnm2-/- pups were born alive. The Cnnm2-/- pups had a significantly lower serum Mg2+ concentration compared to wildtype littermates. Subsequently, adult Cnnm2+/- mice were fed with low, control, or high Mg2+ diets for two weeks. Adult Cnnm2+/- mice showed mild hypomagnesaemia compared to Cnnm2+/+ mice and increased serum Ca2+ levels, independent of dietary Mg2+ intake. Faecal analysis displayed increased Mg2+ and Ca2+ excretion in the Cnnm2+/- mice. Transcriptional profiling of Trpm6, Trpm7, and Slc41a1 in kidneys and colon did not reveal effects based on genotype. Microcomputed tomography analysis of the femurs demonstrated equal bone morphology and density. In conclusion, CNNM2 is vital for embryonic development and Mg2+ homeostasis. Our data suggest a previously undescribed role of CNNM2 in the intestine, which may contribute to the Mg2+ deficiency in mice and patients.
Collapse
|
28
|
Franken GAC, Müller D, Mignot C, Keren B, Lévy J, Tabet AC, Germanaud D, Tejada MI, Kroes HY, Nievelstein RAJ, Brimble E, Ruzhnikov M, Claverie-Martin F, Szczepańska M, Ćuk M, Latta F, Konrad M, Martínez-Cruz LA, Bindels RJM, Hoenderop JGJ, Schlingmann KP, de Baaij JHF. The phenotypic and genetic spectrum of patients with heterozygous mutations in cyclin M2 (CNNM2). Hum Mutat 2021; 42:473-486. [PMID: 33600043 PMCID: PMC8248058 DOI: 10.1002/humu.24182] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/21/2022]
Abstract
Hypomagnesemia, seizures, and intellectual disability (HSMR) syndrome is a rare disorder caused by mutations in the cyclin M2 (CNNM2) gene. Due to the limited number of cases, extensive phenotype analyses of these patients have not been performed, hindering early recognition of patients. In this study, we established the largest cohort of HSMR to date, aiming to improve recognition and diagnosis of this complex disorder. Eleven novel variants in CNNM2 were identified in nine single sporadic cases and in two families with suspected HSMR syndrome. 25Mg2+ uptake assays demonstrated loss‐of‐function in seven out of nine variants in CNNM2. Interestingly, the pathogenic mutations resulted in decreased plasma membrane expression. The phenotype of those affected by pathogenic CNNM2 mutations was compared with five previously reported cases of HSMR. All patients suffered from hypomagnesemia (0.44–0.72 mmol/L), which could not be fully corrected by Mg2+ supplementation. The majority of patients (77%) experienced generalized seizures and exhibited mild to moderate intellectual disability and speech delay. Moreover, severe obesity was present in most patients (89%). Our data establish hypomagnesemia, seizures, intellectual disability, and obesity as hallmarks of HSMR syndrome. The assessment of these major features offers a straightforward tool for the clinical diagnosis of HSMR.
Collapse
Affiliation(s)
- Gijs A C Franken
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolism, Charité Universitäts Medizin, Berlin, Germany
| | - Cyril Mignot
- Département de Genetique, Centre de Référence Déficiences Intellectuelles de Causes Rares, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Boris Keren
- Département de Génétique, Groupe Hospitalier, Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jonathan Lévy
- Genetics Department, AP-HP, Robert-Debré University Hospital, Paris, France
| | - Anne-Claude Tabet
- Genetics Department, AP-HP, Robert-Debré University Hospital, Paris, France
| | - David Germanaud
- Pediatric Neurology Department, Centre de Référence Déficiences Intellectuelles de Causes Rares, Service de Neurologie Pédiatrique, Hôpital Robert-Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - María-Isabel Tejada
- Osakidetza Basque Health Service, Cruces University Hospital, Genetics Service and Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Spanish Consortium for Research on Rare Diseases (CIBERER), Valencia, Spain
| | - Hester Y Kroes
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rutger A J Nievelstein
- Department of Pediatric Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elise Brimble
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, California, USA
| | - Maria Ruzhnikov
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, California, USA
| | - Felix Claverie-Martin
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Maria Szczepańska
- Department of Pediatrics, Medical University of Silesia, Katowice, Poland
| | - Martin Ćuk
- Department of Pediatrics, Children's Hospital Zagreb, Zagreb, Croatia
| | - Femke Latta
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Konrad
- Department of General Pediatrics, University Children's Hospital, Münster, Germany
| | - Luis A Martínez-Cruz
- Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Technology Park of Bizkaia, Derio, Spain
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - 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
|
29
|
Franken GAC, Adella A, Bindels RJM, de Baaij JHF. Mechanisms coupling sodium and magnesium reabsorption in the distal convoluted tubule of the kidney. Acta Physiol (Oxf) 2021; 231:e13528. [PMID: 32603001 PMCID: PMC7816272 DOI: 10.1111/apha.13528] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/29/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Hypomagnesaemia is a common feature of renal Na+ wasting disorders such as Gitelman and EAST/SeSAME syndrome. These genetic defects specifically affect Na+ reabsorption in the distal convoluted tubule, where Mg2+ reabsorption is tightly regulated. Apical uptake via TRPM6 Mg2+ channels and basolateral Mg2+ extrusion via a putative Na+ -Mg2+ exchanger determines Mg2+ reabsorption in the distal convoluted tubule. However, the mechanisms that explain the high incidence of hypomagnesaemia in patients with Na+ wasting disorders of the distal convoluted tubule are largely unknown. In this review, we describe three potential mechanisms by which Mg2+ reabsorption in the distal convoluted tubule is linked to Na+ reabsorption. First, decreased activity of the thiazide-sensitive Na+ /Cl- cotransporter (NCC) results in shortening of the segment, reducing the Mg2+ reabsorption capacity. Second, the activity of TRPM6 and NCC are determined by common regulatory pathways. Secondary effects of NCC dysregulation such as hormonal imbalance, therefore, might disturb TRPM6 expression. Third, the basolateral membrane potential, maintained by the K+ permeability and Na+ -K+ -ATPase activity, provides the driving force for Na+ and Mg2+ extrusion. Depolarisation of the basolateral membrane potential in Na+ wasting disorders of the distal convoluted tubule may therefore lead to reduced activity of the putative Na+ -Mg2+ exchanger SLC41A1. Elucidating the interconnections between Mg2+ and Na+ transport in the distal convoluted tubule is hampered by the currently available models. Our analysis indicates that the coupling of Na+ and Mg2+ reabsorption may be multifactorial and that advanced experimental models are required to study the molecular mechanisms.
Collapse
Affiliation(s)
- Gijs A. C. Franken
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - Anastasia Adella
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - René J. M. Bindels
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| | - Jeroen H. F. de Baaij
- Department of PhysiologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenthe Netherlands
| |
Collapse
|
30
|
Laursen WJ, Garrity PA. A divalent boost from magnesium. eLife 2021; 10:65359. [PMID: 33504427 PMCID: PMC7843128 DOI: 10.7554/elife.65359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 11/13/2022] Open
Abstract
Enhanced levels of dietary magnesium improve long-term memory in fruit flies.
Collapse
Affiliation(s)
- Willem J Laursen
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, United States
| | - Paul A Garrity
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, United States
| |
Collapse
|
31
|
Mg 2+ Transporters in Digestive Cancers. Nutrients 2021; 13:nu13010210. [PMID: 33450887 PMCID: PMC7828344 DOI: 10.3390/nu13010210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/08/2023] Open
Abstract
Despite magnesium (Mg2+) representing the second most abundant cation in the cell, its role in cellular physiology and pathology is far from being elucidated. Mg2+ homeostasis is regulated by Mg2+ transporters including Mitochondrial RNA Splicing Protein 2 (MRS2), Transient Receptor Potential Cation Channel Subfamily M, Member 6/7 (TRPM6/7), Magnesium Transporter 1 (MAGT1), Solute Carrier Family 41 Member 1 (SCL41A1), and Cyclin and CBS Domain Divalent Metal Cation Transport Mediator (CNNM) proteins. Recent data show that Mg2+ transporters may regulate several cancer cell hallmarks. In this review, we describe the expression of Mg2+ transporters in digestive cancers, the most common and deadliest malignancies worldwide. Moreover, Mg2+ transporters’ expression, correlation and impact on patient overall and disease-free survival is analyzed using Genotype Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) datasets. Finally, we discuss the role of these Mg2+ transporters in the regulation of cancer cell fates and oncogenic signaling pathways.
Collapse
|
32
|
Al Alawi AM, Al Badi A, Al Huraizi A, Falhammar H. Magnesium: The recent research and developments. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 96:193-218. [PMID: 34112353 DOI: 10.1016/bs.afnr.2021.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Magnesium is the fourth most abundant mineral in the human body, which facilitates more than 300 enzymatic reactions. Magnesium is essential for nucleic material and protein synthesis, neuromuscular conduction, cardiac contractility, energy metabolism, and immune system function. Gastrointestinal system and kidneys closely regulate magnesium absorption and elimination to maintain adequate storage of magnesium. Magnesium deficiency has been linked to many diseases and poor health outcomes. Magnesium has also been proven to be an effective therapeutic agent in many diseases, such as bronchial asthma, cardiac arrhythmia, and pre-eclampsia.
Collapse
Affiliation(s)
- Abdullah M Al Alawi
- Department of Medicine, Sultan Qaboos University Hospital, Muscat, Oman; Oman Medical Specialty Board, Muscat, Oman.
| | | | - Aisha Al Huraizi
- Department of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| | - Henrik Falhammar
- Department of Endocrinology, Metabolism, and Diabetes, Karolinska University Hospital, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden; Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| |
Collapse
|
33
|
Wu Y, Funato Y, Meschi E, Jovanoski KD, Miki H, Waddell S. Magnesium efflux from Drosophila Kenyon cells is critical for normal and diet-enhanced long-term memory. eLife 2020; 9:61339. [PMID: 33242000 PMCID: PMC7843133 DOI: 10.7554/elife.61339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Dietary magnesium (Mg2+) supplementation can enhance memory in young and aged rats. Memory-enhancing capacity was largely ascribed to increases in hippocampal synaptic density and elevated expression of the NR2B subunit of the NMDA-type glutamate receptor. Here we show that Mg2+ feeding also enhances long-term memory in Drosophila. Normal and Mg2+-enhanced fly memory appears independent of NMDA receptors in the mushroom body and instead requires expression of a conserved CNNM-type Mg2+-efflux transporter encoded by the unextended (uex) gene. UEX contains a putative cyclic nucleotide-binding homology domain and its mutation separates a vital role for uex from a function in memory. Moreover, UEX localization in mushroom body Kenyon cells (KCs) is altered in memory-defective flies harboring mutations in cAMP-related genes. Functional imaging suggests that UEX-dependent efflux is required for slow rhythmic maintenance of KC Mg2+. We propose that regulated neuronal Mg2+ efflux is critical for normal and Mg2+-enhanced memory. The proverbial saying ‘you are what you eat’ perfectly summarizes the concept that our diet can influence both our mental and physical health. We know that foods that are good for the heart, such as nuts, oily fish and berries, are also good for the brain. We know too that vitamins and minerals are essential for overall good health. But is there any evidence that increasing your intake of specific vitamins or minerals could help boost your brain power? While it might sound almost too good to be true, there is some evidence that this is the case for at least one mineral, magnesium. Studies in rodents have shown that adding magnesium supplements to food improves how well the animals perform on memory tasks. Both young and old animals benefit from additional magnesium. Even elderly rodents with a condition similar to Alzheimer’s disease show less memory loss when given magnesium supplements. But what about other species? Wu et al. now show that magnesium supplements also boost memory performance in fruit flies. One group of flies was fed with standard cornmeal for several days, while the other group received cornmeal supplemented with magnesium. Both groups were then trained to associate an odor with a food reward. Flies that had received the extra magnesium showed better memory for the odor when tested 24 hours after training. Wu et al. show that magnesium improves memory in the flies via a different mechanism to that reported previously for rodents. In rodents, magnesium increased levels of a receptor protein for a brain chemical called glutamate. In fruit flies, by contrast, the memory boost depended on a protein that transports magnesium out of neurons. Mutant flies that lacked this transporter showed memory impairments. Unlike normal flies, those without the transporter showed no memory improvement after eating magnesium-enriched food. The results suggest that the transporter may help adjust magnesium levels inside brain cells in response to neural activity. Humans produce four variants of this magnesium transporter, each encoded by a different gene. One of these transporters has already been implicated in brain development. The findings of Wu et al. suggest that the transporters may also act in the adult brain to influence cognition. Further studies are needed to test whether targeting the magnesium transporter could ultimately hold promise for treating memory impairments.
Collapse
Affiliation(s)
- Yanying Wu
- Centre for Neural Circuits and Behaviour, The University of Oxford, Tinsley Building, Oxford, United Kingdom
| | - Yosuke Funato
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Eleonora Meschi
- Centre for Neural Circuits and Behaviour, The University of Oxford, Tinsley Building, Oxford, United Kingdom
| | - Kristijan D Jovanoski
- Centre for Neural Circuits and Behaviour, The University of Oxford, Tinsley Building, Oxford, United Kingdom
| | - Hiroaki Miki
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, The University of Oxford, Tinsley Building, Oxford, United Kingdom
| |
Collapse
|
34
|
García-Castaño A, Madariaga L, Antón-Gamero M, Mejia N, Ponce J, Gómez-Conde S, Pérez de Nanclares G, De la Hoz AB, Martínez R, Saso L, Martínez de LaPiscina I, Urrutia I, Velasco O, Aguayo A, Castaño L, Gaztambide S. Novel variant in the CNNM2 gene associated with dominant hypomagnesemia. PLoS One 2020; 15:e0239965. [PMID: 32997713 PMCID: PMC7527205 DOI: 10.1371/journal.pone.0239965] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
The maintenance of magnesium (Mg2+) homeostasis is essential for human life. The Cystathionine-β-synthase (CBS)-pair domain divalent metal cation transport mediators (CNNMs) have been described to be involved in maintaining Mg2+ homeostasis. Among these CNNMs, CNNM2 is expressed in the basolateral membrane of the kidney tubules where it is involved in Mg2+ reabsorption. A total of four patients, two of them with a suspected disorder of calcium metabolism, and two patients with a clinical diagnosis of primary tubulopathy were screened for mutations by Next-Generation Sequencing (NGS). We found one novel likely pathogenic variant in the heterozygous state (c.2384C>A; p.(Ser795*)) in the CNNM2 gene in a family with a suspected disorder of calcium metabolism. In this family, hypomagnesemia was indirectly discovered. Moreover, we observed three novel variants of uncertain significance in heterozygous state in the other three patients (c.557G>C; p.(Ser186Thr), c.778A>T; p.(Ile260Phe), and c.1003G>A; p.(Asp335Asn)). Our study shows the utility of Next-Generation Sequencing in unravelling the genetic origin of rare diseases. In clinical practice, serum Mg2+ should be determined in calcium and PTH-related disorders.
Collapse
Affiliation(s)
| | - Leire Madariaga
- Paediatric Nephrology Department, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, CIBERDEM, CIBERER, University of the Basque Country (UPV-EHU), Bizkaia, Spain
| | | | - Natalia Mejia
- Faculty of Medicine, University of Los Andes, Bogotá, Colombia
| | - Jenny Ponce
- Paediatric Department, Hospital Nacional Docente Madre-Niño San Bartolomé, Lima, Peru
| | | | - Gustavo Pérez de Nanclares
- Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, CIBERDEM, CIBERER, Bizkaia, Spain
| | | | - Rosa Martínez
- Biocruces Bizkaia Health Research Institute, CIBERDEM, CIBERER, Bizkaia, Spain
| | - Laura Saso
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | | | - Inés Urrutia
- Biocruces Bizkaia Health Research Institute, CIBERDEM, CIBERER, Bizkaia, Spain
| | - Olaia Velasco
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | - Aníbal Aguayo
- Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, CIBERDEM, CIBERER, Bizkaia, Spain
| | - Luis Castaño
- Endocrinology and Nutrition Department, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, CIBERDEM, CIBERER, University of the Basque Country (UPV-EHU), Bizkaia, Spain
| | - Sonia Gaztambide
- Endocrinology and Nutrition Department, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, CIBERDEM, CIBERER, University of the Basque Country (UPV-EHU), Bizkaia, Spain
- * E-mail:
| |
Collapse
|
35
|
Mg2+-ATP Sensing in CNNM, a Putative Magnesium Transporter. Structure 2020; 28:324-335.e4. [DOI: 10.1016/j.str.2019.11.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/27/2019] [Indexed: 01/10/2023]
|
36
|
Giménez-Mascarell P, Oyenarte I, González-Recio I, Fernández-Rodríguez C, Corral-Rodríguez MÁ, Campos-Zarraga I, Simón J, Kostantin E, Hardy S, Díaz Quintana A, Zubillaga Lizeaga M, Merino N, Diercks T, Blanco FJ, Díaz Moreno I, Martínez-Chantar ML, Tremblay ML, Müller D, Siliqi D, Martínez-Cruz LA. Structural Insights into the Intracellular Region of the Human Magnesium Transport Mediator CNNM4. Int J Mol Sci 2019; 20:E6279. [PMID: 31842432 PMCID: PMC6940986 DOI: 10.3390/ijms20246279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
The four member family of "Cyclin and Cystathionine β-synthase (CBS) domain divalent metal cation transport mediators", CNNMs, are the least-studied mammalian magnesium transport mediators. CNNM4 is abundant in the brain and the intestinal tract, and its abnormal activity causes Jalili Syndrome. Recent findings show that suppression of CNNM4 in mice promotes malignant progression of intestinal polyps and is linked to infertility. The association of CNNM4 with phosphatases of the regenerating liver, PRLs, abrogates its Mg2+-efflux capacity, thus resulting in an increased intracellular Mg2+ concentration that favors tumor growth. Here we present the crystal structures of the two independent intracellular domains of human CNNM4, i.e., the Bateman module and the cyclic nucleotide binding-like domain (cNMP). We also derive a model structure for the full intracellular region in the absence and presence of MgATP and the oncogenic interacting partner, PRL-1. We find that only the Bateman module interacts with ATP and Mg2+, at non-overlapping sites facilitating their positive cooperativity. Furthermore, both domains dimerize autonomously, where the cNMP domain dimer forms a rigid cleft to restrict the Mg2+ induced sliding of the inserting CBS1 motives of the Bateman module, from a twisted to a flat disk shaped dimer.
Collapse
Grants
- ETORTEK IE05-147 Departamento de Industria, Innovación, Comercio y Turismo del Gobierno Vasco
- IE07-202 Departamento de Industria, Innovación, Comercio y Turismo del Gobierno Vasco
- 7/13/08/2006/11 Diputación Foral de Bizkaia
- 7/13/08/2005/14 Diputación Foral de Bizkaia
- BFU2010-17857 Ministerio de Ciencia e Innovación
- BFU2013-47531-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- BES-2014-068464 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- BFU2016-77408-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- BES-2017-080435 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- CSD2008-00005 MICINN CONSOLIDER-INGENIO 2010 Program
- BAG MX20113 Diamond Light source
- 2013111114 Gobierno Vasco-Departamento de Salud
- SAF2017-87301-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- BIO15/CA/014 EITB Maratoia
- SEV-2016-0644 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- 12.01.134/2bT4 Berlin Institute of Health
- #343439 Canadian Institute for Health Research
- MX15832-9 Diamond Light Source
- MX15832-10 Diamond Light Source
- PGC2018-096049-B100 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- CTQ2017-83810-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- PI2010-17 Departamento de Educación, Universidades e Investigación del Gobierno Vasco
- BAG 2019073624 ALBA Synchrotron
Collapse
Affiliation(s)
- Paula Giménez-Mascarell
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Iker Oyenarte
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Irene González-Recio
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Carmen Fernández-Rodríguez
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - María Ángeles Corral-Rodríguez
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Igone Campos-Zarraga
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Jorge Simón
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| | - Elie Kostantin
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (E.K.); (S.H.); (M.L.T.)
| | - Serge Hardy
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (E.K.); (S.H.); (M.L.T.)
| | - Antonio Díaz Quintana
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla—CSIC. Avda. Americo Vespucio 49, 41092 Sevilla, Spain; (A.D.Q.); (I.D.M.)
| | - Mara Zubillaga Lizeaga
- Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 800, 48160 Derio, Spain; (M.Z.L.); (N.M.); (T.D.); (F.J.B.)
| | - Nekane Merino
- Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 800, 48160 Derio, Spain; (M.Z.L.); (N.M.); (T.D.); (F.J.B.)
| | - Tammo Diercks
- Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 800, 48160 Derio, Spain; (M.Z.L.); (N.M.); (T.D.); (F.J.B.)
| | - Francisco J. Blanco
- Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 800, 48160 Derio, Spain; (M.Z.L.); (N.M.); (T.D.); (F.J.B.)
- IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, 48013 Bilbao, Spain
| | - Irene Díaz Moreno
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla—CSIC. Avda. Americo Vespucio 49, 41092 Sevilla, Spain; (A.D.Q.); (I.D.M.)
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Spain
| | - Michel L. Tremblay
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (E.K.); (S.H.); (M.L.T.)
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Disorders, Charité Universitäts medizin, 13353 Berlin, Germany;
| | - Dritan Siliqi
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche (CNR), Via G. Amendola 122/O, 70126 Bari, Italy;
| | - Luis Alfonso Martínez-Cruz
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Bizkaia Science and Technology Park Bld 801A, 48160 Derio, Spain; (P.G.-M.); (I.O.); (I.G.-R.); (C.F.-R.); (M.Á.C.-R.); (I.C.-Z.); (J.S.); (M.L.M.-C.)
| |
Collapse
|
37
|
Trapani V, Wolf FI. Dysregulation of Mg2+ homeostasis contributes to acquisition of cancer hallmarks. Cell Calcium 2019; 83:102078. [DOI: 10.1016/j.ceca.2019.102078] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/26/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
|
38
|
Trachsel E, Redder P, Linder P, Armitano J. Genetic screens reveal novel major and minor players in magnesium homeostasis of Staphylococcus aureus. PLoS Genet 2019; 15:e1008336. [PMID: 31415562 PMCID: PMC6711546 DOI: 10.1371/journal.pgen.1008336] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/27/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022] Open
Abstract
Magnesium is one of the most abundant metal ions in living cells. Very specific and devoted transporters have evolved for transporting Mg2+ ions across the membrane and maintain magnesium homeostasis. Using genetic screens, we were able to identify the main players in magnesium homeostasis in the opportunistic pathogen Staphylococcus aureus. Here, we show that import of magnesium relies on the redundant activity of either CorA2 or MgtE since in absence of these two importers, bacteria require increased amounts of magnesium in the medium. A third CorA-like importer seems to play a minor role, at least under laboratory conditions. For export of magnesium, we identified two proteins, MpfA and MpfB. MpfA, is the main actor since it is essential for growth in high magnesium concentrations. We show that gain of function mutations or overexpression of the minor factor, MpfB, which is part of a sigmaB controlled stress response regulon, can compensate for the absence of MpfA.
Collapse
Affiliation(s)
- Emilie Trachsel
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Redder
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- LMGM UMR5100, Centre de Biologie Integrative, Paul Sabatier University, Toulouse, France
| | - Patrick Linder
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Joshua Armitano
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
39
|
Magnesium-sensitive upstream ORF controls PRL phosphatase expression to mediate energy metabolism. Proc Natl Acad Sci U S A 2019; 116:2925-2934. [PMID: 30718434 DOI: 10.1073/pnas.1815361116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Phosphatases of regenerating liver (PRL-1, PRL-2, and PRL-3, also known as PTP4A1, PTP4A2, and PTP4A3) control magnesium homeostasis through an association with the CNNM magnesium transport regulators. Although high PRL levels have been linked to cancer progression, regulation of their expression is poorly understood. Here we show that modulating intracellular magnesium levels correlates with a rapid change of PRL expression by a mechanism involving its 5'UTR mRNA region. Mutations or CRISPR-Cas9 targeting of the conserved upstream ORF present in the mRNA leader derepress PRL protein synthesis and attenuate the translational response to magnesium levels. Mechanistically, magnesium depletion reduces intracellular ATP but up-regulates PRL protein expression via activation of the AMPK/mTORC2 pathway, which controls cellular energy status. Hence, altered PRL-2 expression leads to metabolic reprogramming of the cells. These findings uncover a magnesium-sensitive mechanism controlling PRL expression, which plays a role in cellular bioenergetics.
Collapse
|
40
|
Chen YS, Kozlov G, Fakih R, Funato Y, Miki H, Gehring K. The cyclic nucleotide-binding homology domain of the integral membrane protein CNNM mediates dimerization and is required for Mg 2+ efflux activity. J Biol Chem 2018; 293:19998-20007. [PMID: 30341174 PMCID: PMC6311497 DOI: 10.1074/jbc.ra118.005672] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/16/2018] [Indexed: 01/07/2023] Open
Abstract
Proteins of the cyclin M family (CNNMs; also called ancient conserved domain proteins, or ACDPs) are represented by four integral membrane proteins that have been proposed to function as Mg2+ transporters. CNNMs are associated with a number of genetic diseases affecting ion movement and cancer via their association with highly oncogenic phosphatases of regenerating liver (PRLs). Structurally, CNNMs contain an N-terminal extracellular domain, a transmembrane domain (DUF21), and a large cytosolic region containing a cystathionine-β-synthase (CBS) domain and a putative cyclic nucleotide-binding homology (CNBH) domain. Although the CBS domain has been extensively characterized, little is known about the CNBH domain. Here, we determined the first crystal structures of the CNBH domains of CNNM2 and CNNM3 at 2.6 and 1.9 Å resolutions. Contrary to expectation, these domains did not bind cyclic nucleotides, but mediated dimerization both in crystals and in solution. Analytical ultracentrifugation experiments revealed an inverse correlation between the propensity of the CNBH domains to dimerize and the ability of CNNMs to mediate Mg2+ efflux. CNBH domains from active family members were observed as both dimers and monomers, whereas the inactive member, CNNM3, was observed only as a dimer. Mutational analysis revealed that the CNBH domain was required for Mg2+ efflux activity of CNNM4. This work provides a structural basis for understanding the function of CNNM proteins in Mg2+ transport and associated diseases.
Collapse
Affiliation(s)
- Yu Seby Chen
- From the Department of Biochemistry and Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada and
| | - Guennadi Kozlov
- From the Department of Biochemistry and Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada and
| | - Rayan Fakih
- From the Department of Biochemistry and Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada and
| | - Yosuke Funato
- the Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroaki Miki
- the Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kalle Gehring
- From the Department of Biochemistry and Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada and , To whom correspondence should be addressed:
Dept. of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Rm. 469, Montreal, Quebec H3G 0B1, Canada. Tel.:
514-398-7287; E-mail:
| |
Collapse
|
41
|
Molecular function and biological importance of CNNM family Mg2+ transporters. J Biochem 2018; 165:219-225. [DOI: 10.1093/jb/mvy095] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022] Open
|
42
|
Accogli A, Scala M, Calcagno A, Napoli F, Di Iorgi N, Arrigo S, Mancardi MM, Prato G, Pisciotta L, Nagel M, Severino M, Capra V. CNNM2 homozygous mutations cause severe refractory hypomagnesemia, epileptic encephalopathy and brain malformations. Eur J Med Genet 2018; 62:198-203. [PMID: 30026055 DOI: 10.1016/j.ejmg.2018.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/01/2018] [Accepted: 07/14/2018] [Indexed: 12/19/2022]
Abstract
Magnesium (Mg2+) plays a crucial role in many biological processes especially in the brain, heart and skeletal muscle. Mg2+ homeostasis is regulated by intestinal absorption and renal reabsorption, involving a combination of different epithelial transport pathways. Mutations in any of these transporters result in hypomagnesemia with variable clinical presentations. Among these, CNNM2 is found along the basolateral membrane of distal tubular segments where it is involved in Mg2+ reabsorption. To date, heterozygous mutations in CNNM2 have been associated with a variable phenotype, ranging from isolated hypomagnesemia to intellectual disability and epilepsy. The only homozygous mutation reported so far, is responsible for hypomagnesemia associated with a severe neurological phenotype characterized by refractory epilepsy, microcephaly, severe global developmental delay and intellectual disability. Here, we report the second homozygous CNNM2 mutation (c.1642G > A,p.Val548Met) in a Moroccan patient, presenting with hypomagnesemia and severe epileptic encephalopathy. Thus, we review and discuss the phenotypic spectrum associated with CNNM2 mutations.
Collapse
Affiliation(s)
- Andrea Accogli
- UOC Neurochirurgia, Istituto Giannina Gaslini, Genova, Italy; Università degli studi di Genova, Italy
| | - Marcello Scala
- UOC Neurochirurgia, Istituto Giannina Gaslini, Genova, Italy; Università degli studi di Genova, Italy
| | | | - Flavia Napoli
- UOC Clinica Pediatrica, Istituto Giannina Gaslini, Genova, Italy
| | - Natascia Di Iorgi
- Università degli studi di Genova, Italy; UOC Clinica Pediatrica, Istituto Giannina Gaslini, Genova, Italy
| | - Serena Arrigo
- UOC Gastroenterologia and Endoscopia Pediatrica, Istituto Giannina Gaslini, Genova, Italy
| | | | - Giulia Prato
- UOC Neuropsichiatria Infantile - Centro Epilessia, Istituto Giannina Gaslini, Genova, Italy
| | - Livia Pisciotta
- Università degli studi di Genova, Italy; UOC Neuropsichiatria Infantile - Centro Epilessia, Istituto Giannina Gaslini, Genova, Italy
| | - Mato Nagel
- Center for Nephrology and Metabolic Disorders, Weisswasser, Germany
| | | | - Valeria Capra
- UOC Neurochirurgia, Istituto Giannina Gaslini, Genova, Italy.
| |
Collapse
|
43
|
Hardy S, Kostantin E, Hatzihristidis T, Zolotarov Y, Uetani N, Tremblay ML. Physiological and oncogenic roles of thePRLphosphatases. FEBS J 2018; 285:3886-3908. [DOI: 10.1111/febs.14503] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/30/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Serge Hardy
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
| | - Elie Kostantin
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
| | - Teri Hatzihristidis
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Medicine Division of Experimental Medicine McGill University Montreal Canada
| | - Yevgen Zolotarov
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
| | - Noriko Uetani
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
| | - Michel L. Tremblay
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
- Department of Medicine Division of Experimental Medicine McGill University Montreal Canada
| |
Collapse
|
44
|
Funato Y, Furutani K, Kurachi Y, Miki H. Rebuttal from Yosuke Funato, Kazuharu Furutani, Yoshihisa Kurachi and Hiroaki Miki. J Physiol 2018; 596:751. [PMID: 29383723 PMCID: PMC5830423 DOI: 10.1113/jp275706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Yosuke Funato
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversitySuitaOsaka565‐0871Japan
| | - Kazuharu Furutani
- Department of PharmacologyGraduate School of MedicineOsaka UniversitySuitaOsaka565‐0871Japan
- Department of Physiology and Membrane BiologyUniversity of CaliforniaDavisCA95616USA
| | - Yoshihisa Kurachi
- Department of PharmacologyGraduate School of MedicineOsaka UniversitySuitaOsaka565‐0871Japan
| | - Hiroaki Miki
- Department of Cellular RegulationResearch Institute for Microbial Diseases, Osaka UniversitySuitaOsaka565‐0871Japan
| |
Collapse
|